Quick viewing(Text Mode)

Proteome Profile of Peripheral Myelin in Healthy Mice and in a Neuropathy

Proteome Profile of Peripheral Myelin in Healthy Mice and in a Neuropathy

TOOLS AND RESOURCES

Proteome profile of peripheral in healthy mice and in a neuropathy model Sophie B Siems1†, Olaf Jahn2†, Maria A Eichel1, Nirmal Kannaiyan3, Lai Man N Wu4, Diane L Sherman4, Kathrin Kusch1, Do¨ rte Hesse2, Ramona B Jung1, Robert Fledrich1,5, Michael W Sereda1,6, Moritz J Rossner3, Peter J Brophy4, Hauke B Werner1*

1Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Go¨ ttingen, Germany; 2Proteomics Group, Max Planck Institute of Experimental Medicine, Go¨ ttingen, Germany; 3Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany; 4Centre for Discovery Sciences, University of Edinburgh, Edinburgh, United Kingdom; 5Institute of Anatomy, University of Leipzig, Leipzig, Germany; 6Department of Clinical Neurophysiology, University Medical Center, Go¨ ttingen, Germany

Abstract Proteome and transcriptome analyses aim at comprehending the molecular profiles of the brain, its -types and subcellular compartments including myelin. Despite the relevance of the peripheral for normal sensory and motor capabilities, analogous approaches to peripheral and peripheral myelin have fallen behind evolving technical standards. Here we assess the peripheral myelin proteome by gel-free, label-free mass-spectrometry for deep quantitative coverage. Integration with RNA--based developmental mRNA-abundance profiles and neuropathy disease illustrates the utility of this resource. Notably, the periaxin- deficient mouse model of the neuropathy Charcot-Marie-Tooth 4F displays a highly pathological myelin proteome profile, exemplified by the discovery of reduced levels of the monocarboxylate *For correspondence: transporter MCT1/SLC16A1 as a novel facet of the neuropathology. This work provides the most [email protected] comprehensive proteome resource thus far to approach development, function and pathology of †These authors contributed peripheral myelin, and a straightforward, accurate and sensitive workflow to address myelin equally to this work diversity in health and disease. Competing interests: The authors declare that no competing interests exist. Introduction Funding: See page 22 The ensheathment of with myelin enables rapid impulse propagation, a prerequisite for nor- Received: 27 August 2019 mal motor and sensory capabilities of vertebrates (Weil et al., 2018; Hartline and Colman, 2007). Accepted: 19 February 2020 This is illustrated by demyelinating neuropathies of the Charcot-Marie-Tooth (CMT) spectrum, in Published: 04 March 2020 which affecting myelin genes as MPZ, PMP22, GJB1 and PRX impair myelin integrity and reduce the velocity of conduction in the peripheral nervous system (PNS) (Rossor et al., Reviewing editor: Beth Stevens, Boston Children’s Hospital, 2013). Developmentally, myelination by Schwann cells in peripheral nerves is regulated by axonal United States neuregulin-1 (Michailov et al., 2004; Taveggia et al., 2005) and the (Chernousov et al., 2008; Petersen et al., 2015; Ghidinelli et al., 2017) that is molecularly linked Copyright Siems et al. This to the abaxonal membrane via and the complex article is distributed under the (Sherman et al., 2001; Masaki et al., 2002; Nodari et al., 2008; Raasakka et al., 2019). In adult- terms of the Creative Commons Attribution License, which hood, the basal lamina continues to enclose all /myelin-units (Hess and Lansing, 1953), proba- permits unrestricted use and bly to maintain myelin. Beyond regulation by extracellular cues, myelination involves multiple redistribution provided that the mediating radial sorting of axons out of Remak bundles, myelin membrane growth and original author and source are layer compaction (Sherman and Brophy, 2005; Pereira et al., 2012; Grove and Brophy, 2014; credited. Monk et al., 2015; Feltri et al., 2016). For example, the Ig-domain containing myelin zero

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 1 of 31 Tools and resources Neuroscience (MPZ; also termed P0) mediates adhesion between adjacent extracellular membrane surfaces in compact myelin (Giese et al., 1992). At their intracellular surfaces, myelin membranes are com- pacted by the cytosolic domain of MPZ/P0 together with (MBP; previously termed P1) (Martini et al., 1995; Nawaz et al., 2013). Not surprisingly, MPZ/P0 and MBP were early identified as the most abundant peripheral myelin proteins (Greenfield et al., 1973; Brostoff et al., 1975). A system of cytoplasmic channels through the otherwise compacted myelin sheath remains non- compacted throughout life, that is the adaxonal myelin layer, paranodal loops, Schmidt-Lanterman incisures (SLI), and abaxonal longitudinal and transverse bands of termed bands of Cajal (Sherman and Brophy, 2005; Nave and Werner, 2014; Kleopa and Sargiannidou, 2015). Non- compacted myelin comprises cytoplasm, cytoskeletal elements, vesicles and -modifying , and thus numerous proteins involved in maintaining the myelin sheath. The cytosolic chan- nels probably also represent transport routes toward Schwann cell-dependent metabolic support of myelinated axons (Court et al., 2004; Beirowski et al., 2014; Dome`nech-Este´vez et al., 2015; Kim et al., 2016; Gonc¸alves et al., 2017; Stassart et al., 2018). Considering that Schwann cells constitute a major proportion of the cells in the PNS, oligonucleo- tide microarray analyses have been used for mRNA abundance profiling of total sciatic nerves (Nagarajan et al., 2002; Le et al., 2005). Indeed, these systematic approaches allowed the identifi- cation of novel myelin constituents including non-compact myelin-associated protein (NCMAP/ MP11) (Ryu et al., 2008). Notwithstanding that the number of known peripheral myelin proteins has grown in recent years, a comprehensive molecular inventory has been difficult to achieve because applications of systematic (‘omics’) approaches specifically to Schwann cells and peripheral myelin remained comparatively scarce, different from studies addressing and CNS myelin (Zhang et al., 2014; Patzig et al., 2016b; Sharma et al., 2015; Thakurela et al., 2016; Marques et al., 2016; de Monasterio-Schrader et al., 2012). One main reason may be that the available techniques were not sufficiently straightforward for general application. For example, the protein composition of peripheral myelin was previously assessed by proteome analysis (Patzig et al., 2011). However, at that time the workflow of sample preparation and data acquisition (schematically depicted in Figure 1A) was very labor-intense and required a substantial amount of input material; yet the depth of the resulting datasets remained limited. In particular, differential myelin proteome analysis by 2-dimensional fluorescence intensity gel electrophoresis (2D-DIGE) requires considerable hands-on-time and technical expertise (Patzig et al., 2011; Kangas et al., 2016). While this method is powerful for the separation of proteoforms (Kusch et al., 2017), it typi- cally suffers from under-representation of highly basic and transmembrane proteins. It thus allows comparing the abundance of only few myelin proteins rather than quantitatively covering the entire myelin proteome. Because of these limitations and an only modest sample-to-sample reproducibility, 2D-DIGE analysis of myelin, although unbiased, has not been commonly applied beyond specialized laboratories. The aim of the present study was to establish a straightforward and readily applicable workflow to facilitate both comprehensive knowledge about the protein composition of peripheral myelin and systematic assessment of differences between two states, for example, pathological alterations in a neuropathy model. The major prerequisites were the biochemical purification of myelin, its solubiliza- tion with the detergent ASB-14 and the subsequent automated digestion with trypsin during filter- aided sample preparation (FASP). The tryptic were fractionated by liquid chromatography and analyzed by mass spectrometry for gel-free, label-free quantitative proteome analysis. More specifically, we used nano-flow ultra-performance liquid chromatography (nanoUPLC) coupled to an electrospray-ionization quadrupole time-of-flight (ESI-QTOF) mass spectrometer with ion mobility option, providing an orthogonal dimension of separation. The utilized data-independent acquisition (DIA) strategy relies on collecting data in an alternating low and elevated energy mode (MSE); it enables simultaneous sequencing and quantification of all peptides entering the mass spec- trometer without prior precursor selection, as reviewed in Neilson et al. (2011) and Distler et al. (2014a). With their high-duty cycle utilized for the acquisition of precursor ions, MSE-type methods are ideally suited to reliably quantify proteins based on peptide intensities. Notably, these methods do not involve the use of spectral libraries in the identification of proteins, different from other DIA strategies. Instead, the achieved high-complexity fragmentation spectra are deconvoluted before submission to dedicated search engines for peptide and protein identification (Geromanos et al.,

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 2 of 31 Tools and resources Neuroscience

A ^ĂŵƉůĞƉƌĞƉĂƌĂƟŽŶ WƌŽƚĞŝŶƐĞƉĂƌĂƟŽŶ dƌLJƉƐŝŶĚŝŐĞƐƟŽŶ WĞƉƟĚĞƐĞƉĂƌĂƟŽŶ DĂƐƐƐƉĞĐƚƌŽŵĞƚĞƌ ĂƚĂĂĐƋƵŝƐŝƟŽŶ Analysis output

WƌĞĐŝƉŝƚĂƟŽŶ 2D-DIGE 'ĞůͲĂŶĚůĂďĞůͲďĂƐĞĚ PMF ϮͲĚŝŵĞŶƐŝŽŶĂůĚŝīĞƌĞŶƟĂů ƵƚŽŵĂƚĞĚŝŶͲŐĞů MALDI-TOF/TOF ĚŝīĞƌĞŶƟĂůĂŶĂůLJƐŝƐ ŇƵŽƌĞƐĐĞŶĐĞŝŶƚĞŶƐŝƚLJŐĞů MS/MS ŝŶĚŝǀŝĚƵĂůƉƌŽƚĞŝŶ ^ŽůƵďŝůŝnjĂƟŽŶ ĞůĞĐƚƌŽƉŚŽƌĞƐŝƐ ASB-14/urea ŝĚĞŶƟĮĐĂƟŽŶ Patzig et al., 2011 WƌĞĐŝƉŝƚĂƟŽŶ YƵĂŶƟĮĐĂƟŽŶŽĨ E DĂŶƵĂůŝŶͲƐŽůƵƟŽŶ nanoUPLC ESI-QTOF MS ĂďƵŶĚĂŶƚƉƌŽƚĞŝŶƐ ^ŽůƵďŝůŝnjĂƟŽŶ 300-400 myelin proteins ĐŝĚͲůĂďŝůĞĚĞƚĞƌŐĞŶƚ Myelin ƉƵƌŝĮĐĂƟŽŶ YƵĂŶƟĮĐĂƟŽŶŽĨ dŚŝƐƐƚƵĚLJ MS E ĂďƵŶĚĂŶƚƉƌŽƚĞŝŶƐ 300-400 myelin proteins Figure 2

ƵƚŽŵĂƚĞĚ UDMS E ^ŽůƵďŝůŝnjĂƟŽŶ nanoUPLC ESI-QTOF ŽŵƉƌĞŚĞŶƐŝǀĞŝŶǀĞŶƚŽƌLJ Figures 3,4 ŝŶͲƐŽůƵƟŽŶ&^W /ŽŶŵŽďŝůŝƚLJƐĞƉĂƌĂƟŽŶ 700-1000 myelin proteins ASB-14/ urea urea/CHAPS

DRE-UDMS E 'ĞůͲĂŶĚůĂďĞůͲĨƌĞĞ /ŽŶŵŽďŝůŝƚLJƐĞƉĂƌĂƟŽŶ ĚŝīĞƌĞŶƟĂůĂŶĂůLJƐŝƐ Figure 5 LJŶĂŵŝĐƌĂŶŐĞĞŶŚĂŶĐĞŵĞŶƚ 400-700 myelin proteins

DIA

B CD MSE (351) Lysate Myelin DRE-UDMSE (554) 1000000 P0 MSE 503 UDMS E (1078) MBP DRE-UDMSE 25 P0/MPZ E 100000 PRX UDMS 23 229 325 MBP 10000 15 4 15 1000 PMP2 1 ppm 250 E This study (1083) PRX 100 Patzig et al., 2011 (516) 3 Kangas et al., 2016 (19) 250 NEFH 10 11 4 KCNA1 55 648 1 1 VDAC 35 421 0 500 1000 83 EGR2 70 # proteins

Figure 1. Proteome analysis of peripheral myelin. (A) Schematic illustration of a previous approach to the peripheral myelin proteome (Patzig et al., 2011) compared with the present workflow. Note that the current workflow allows largely automated sample processing and omits labor-intense 2- dimensional differential gel-electrophoresis, thereby considerably reducing hands-on time. Nano LC-MS analysis by data-independent acquisition (DIA) using three different data acquisition modes provides efficient identification and quantification of abundant myelin proteins (MSE; see Figure 2), a comprehensive inventory (UDMSE; see Figures 3–4) and gel-free differential analysis of hundreds of distinct proteins (DRE-UDMSE; see Figure 5). Samples were analyzed in three biological replicates. (B) Immunoblot of myelin biochemically enriched from sciatic nerves of wild-type mice at postnatal day 21 (). Equal amounts of corresponding nerve lysate were loaded to compare the abundance of marker proteins for compact myelin (MPZ/P0, MBP, PMP2), non-compact myelin (PRX), the Schwann (KROX20/EGR2), axons (NEFH, KCNA1) and mitochondria (VDAC). Blots show n = 2 biological replicates representative of n = 3 biological replicates. Note that myelin markers are enriched in purified myelin; other cellular markers are reduced. (C) Number and relative abundance of proteins identified in myelin purified from the sciatic nerves of wild-type mice using three different data acquisition modes (MSE, UDMSE, DRE-UDMSE). Note that MSE (orange) provides the best information about the relative abundance of high-abundant myelin proteins (dynamic range of more than four orders of magnitude) but identifies comparatively fewer proteins in purified myelin. UDMSE (blue) identifies the largest number of proteins but provides only a lower dynamic range of about three orders of magnitude. DRE-UDMSE (green) identifies an intermediate number of proteins with an intermediate dynamic range of about four orders of magnitude. Note that MSE with very high dynamic range is required for the quantification of the exceptionally abundant (MPZ/P0), myelin basic protein (MBP) and periaxin (PRX). ppm, parts per million. (D) Venn diagram comparing the number of proteins identified in PNS myelin by MSE, UDMSE and DRE-UDMSE. Note the high overlap of identified proteins. (E) Venn diagram of the proteins identified in PNS myelin by UDMSE in this study compared with those identified in two previous approaches (Patzig et al., 2011; Kangas et al., 2016). The online version of this article includes the following source data and figure supplement(s) for figure 1: Figure 1 continued on next page

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 3 of 31 Tools and resources Neuroscience

Figure 1 continued Source data 1. Label-free quantification of proteins in wild-type PNS myelin fractions by three different data acquisition modes Identification and quan- tification data of detected myelin-associated proteins. Figure supplement 1. Clustered heatmap of Pearson’s correlation coefficients for protein abundance comparing data acquisition modes.

2009; Li et al., 2009). In the MSE mode, this deconvolution involves precursor-fragment ion align- ment solely on the basis of chromatographic elution profiles. On top, drift times of ion mobility-sep- arated precursors are used in the high-definition (HD)MSE mode. An expansion of the latter, referred to as the ultra-definition (UD)MSE mode, additionally implements drift time-dependent colli- sion energy profiles for more effective precursor fragmentation (Distler et al., 2016; Distler et al., 2014b). Indeed, compared to the previously used manual handling and in-gel digestion, the current work- flow (schematically depicted in Figure 1A) is considerably less labor-intense, and automated FASP increases sample-to-sample reproducibility. Moreover, differential analysis by quantitative mass spectrometry (MS) facilitates reproducible quantification of hundreds rather than a few distinct mye- lin proteins. Together, the present workflow increases the efficacy of assessing the peripheral myelin proteome while shifting the main workload from manual sample preparation and gel-separation to automated acquisition and processing of data. We propose that comprehending the expression pro- files of all myelin proteins in the healthy PNS and in myelin-related disorders can contribute to advancing our understanding of the physiology and pathophysiology of peripheral nerves.

Results Purification of peripheral myelin We biochemically enriched myelin as a -weight membrane fraction from pools of sciatic nerves dissected from mice at postnatal day 21 (P21) using an established protocol of discontinuous sucrose density gradient centrifugation (Patzig et al., 2011; Larocca and Norton, 2006), in which myelin membranes accumulate at the interface between 0.29 and 0.85 M sucrose. By immunoblotting, pro- teins specific for both compact (MPZ/P0, MBP, PMP2) and non-compact (PRX) myelin were substan- tially enriched in the myelin fraction compared to nerve lysates (Figure 1B). Conversely, axonal (NEFH, KCNA1) and mitochondrial (VDAC) proteins and a marker for the Schwann cell nucleus (KROX20/EGR2) were strongly reduced in purified myelin. Together, these results imply that bio- chemically purified peripheral myelin is suitable for systematic analysis of its protein composition.

Proteome analysis of peripheral myelin It has long been difficult to accurately quantify the most abundant myelin proteins both in the CNS (PLP, MBP, CNP; Jahn et al., 2009) and the PNS (MPZ/P0, MBP, PRX; this work), probably owing to their exceptionally high relative abundance. For example, the major CNS myelin constituents PLP, MBP and CNP comprise 17, 8 and 4% of the total myelin protein, respectively (Jahn et al., 2009). We have recently provided proof of principle (Erwig et al., 2019a) that the mass spectrometric quantification of these high-abundant myelin proteins is accurate and precise when data are acquired in the MSE data acquisition mode and proteins are quantified according to the TOP3 method, that is if their abundance values are obtained based on the proven correlation between the average intensity of the three peptides exhibiting the most intense mass spectrometry response and the absolute amount of their source protein (Silva et al., 2006; Ahrne´ et al., 2013). Using data acquisition by MSE we confirmed that CNP constitutes about 4% of the total CNS myelin proteome and that the abundance of CNP in myelin from mice heterozygous for the Cnp (CnpWT/null) compared to wild-type mice is 50.7% (±0.4%), in agreement with the halved gene dosage and gel- based quantification by silver staining or immunoblotting (Erwig et al., 2019a). When applying the MSE mode to PNS myelin, we quantified 351 proteins with a false discovery rate (FDR) of <1% at peptide and protein level and an average sequence coverage of 35.5% (Fig- ure 1—source data 1). While MSE (labeled in orange in Figure 1C) indeed provided a dynamic range of more than four orders of magnitude and thus quantitatively covered the exceptionally abundant myelin proteins MPZ/P0, MBP and PRX, the number of quantified proteins appeared

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 4 of 31 Tools and resources Neuroscience limited when spectral complexity was deconvoluted solely on the basis of chromatographic elution profiles. Accordingly, by using the UDMSE mode, which comprises ion mobility for additional pep- tide separation as well as drift time-specific collision energies for peptide fragmentation, proteome coverage was increased about three-fold (1078 proteins quantified; average sequence coverage 34.3%; Figure 1—source data 1). However, the dynamic range of UDMSE (labeled in blue in Figure 1C) was found to be somewhat compressed compared to that of MSE, which can be consid- ered an expectable feature of traveling wave ion mobility devices (Dodds and Baker, 2019), where the analysis of pulsed ion packages leads to a temporal and spatial binning of peptides during ion mobility separation. Indeed, this manifests as a ceiling effect for the detection of exceptionally intense peptide signals, which results in an underestimation of the relative abundance of MPZ/P0, MBP and PRX by UDMSE. The complementary of the MSE and UDMSE data acquisition modes led us to conclude that a comprehensive analysis of the myelin proteome that facilitates both correct quantification of the most abundant proteins and deep quantitative coverage of the proteome would require analyz- ing the same set of samples with two different instrument settings for MSE and UDMSE, respectively. Considering that instrument time is a bottleneck for the routine differential proteome analysis of myelin from mutant mice, we aimed to combine the strengths of MSE and UDMSE into a single data acquisition mode. Based on a enrichment analysis for cellular components of the 200 proteins of highest and lowest abundance from the UDMSE dataset, we realized that the ‘bottom ‘of the quantified proteome is probably largely unrelated to myelin but dominated by contaminants from other subcellular sources including mitochondria. We thus reasoned that for a myelin-directed data acquisition mode, proteome depth may be traded in for a gain in dynamic range and devised a novel method referred to as dynamic range enhancement (DRE)-UDMSE, in which a deflection lens is used to cycle between full and reduced ion transmission during mass spectrometric scanning. Indeed, DRE-UDMSE quantified an intermediate number of proteins in PNS myelin (554 proteins; average sequence coverage 30.6%; Figure 1—source data 1) while providing an intermediate dynamic range (labeled in green in Figure 1C). We thus consider DRE-UDMSE as the data acquisition mode of choice most suitable for routine differential myelin proteome profiling (see below). Overall, we found a high reproducibility between replicates and even among the different data acquisition modes as indicated by Pearson’s correlation coefficients for protein abundance in the range of 0.765–0.997 (Figure 1—figure supplement 1). When comparing the proteins identified in PNS myelin using the three data acquisition modes, we found a very high overlap (Figure 1D). We also found a high overlap (Figure 1E) between the proteins identified in the present study by UDMSE and those detected in previous proteomic approaches to PNS myelin (Patzig et al., 2011; Kangas et al., 2016), thus allowing a high level of confidence. Together, the three data acquisition modes exhibit distinct strengths in the efficient quantification of exceptionally abundant proteins (MSE), establishing a comprehensive inventory (UDMSE) and gel-free, label-free differential analysis of hundreds of distinct proteins (DRE-UDMSE) in peripheral myelin (see Figure 1A). Yet, analyzing the same set of samples by different modes may not always be feasible in all routine applications when considering required instrument time.

Relative abundance of peripheral myelin proteins Considering that MSE provides the high dynamic range required for the quantification of the most abundant myelin proteins, we calculated the relative abundance of the 351 proteins identified in myelin by MSE (Figure 1—source data 1). According to quantitative assessment of this dataset, the most abundant PNS myelin protein, myelin protein zero (MPZ/P0), constitutes 44% (+/- 4% relative standard deviation (RSD)) of the total myelin protein (Figure 2). Myelin basic protein (MBP), periaxin (PRX) and -29 (CD9) constitute 18% (+/- 1% RSD), 15% (+/- 1%) and 1% (+/- 0.2%) of the total myelin protein, respectively (Figure 2). For MPZ/P0 and MBP, our quantification by MSE is in agreement with but specifies prior estimations upon gel-separation and protein labeling by Sudan- Black, Fast-Green or Coomassie-Blue, in which they were judged to constitute 45–70% and 2–26% of the total myelin protein, respectively (Greenfield et al., 1973; Micko and Schlaepfer, 1978; Smith and Curtis, 1979; Whitaker, 1981). However, gel-based estimates of the relative abundance of myelin proteins were not very precise with respect to many other proteins, including those of high molecular weight. Indeed, periaxin was identified as a constituent of peripheral myelin after the advent of gradient SDS-PAGE gels (Gillespie et al., 1994), which allowed improved migration of

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 5 of 31 Tools and resources Neuroscience

Protein % (+/- RSD) Protein % (+/- RSD) P0/ MPZ 44.48 4.15 LAMC1 0.06 0.01 MBP 18.35 1.15 RAC1 0.05 0.01 PRX 15.48 1.46 VCL 0.04 0.00 Others CD9 0.77 0.15 CA2 0.04 0.01 SPTBN1 0.52 0.06 SEPT2 0.04 0.00 SPTAN1 0.42 0.07 CFL1 0.04 0.01 EPB41L2 0.38 0.05 CRYAB 0.03 0.00 VIM 0.34 0.07 MPP6 0.03 0.00 CNP 0.33 0.06 CAV1 0.03 0.00 P0 TF 0.33 0.03 SEPT7 0.02 0.00 PMP2 0.32 0.03 GJC3 0.02 0.00 MAP1B 0.31 0.06 RDX 0.02 0.00 PRX FASN 0.29 0.04 CDC42 0.02 0.01 ANXA2 0.22 0.03 PFN1 0.02 0.00 MAG 0.20 0.02 SEPT11 0.02 0.00 PLP1 0.17 0.02 CRMP1 0.02 0.00 CD81 0.12 0.01 RHOA 0.02 0.00 MBP GSN 0.11 0.02 MAPK3 0.02 0.00 PLLP 0.11 0.01 MAPK1 0.01 0.00 BCAS1 0.09 0.00 CMTM5 0.01 0.00 NDRG1 0.09 0.03 JAM3 0.01 0.00 MSN 0.09 0.01 DYNLL1 0.01 0.03 EPB41L3 0.07 0.01 EZR 0.01 0.00 NID1 0.07 0.01 CD59A 0.01 0.00 CADM4 0.07 0.01 Others 15.39

Figure 2. Relative abundance of peripheral myelin proteins. MSE was used to identify and quantify proteins in myelin purified from the sciatic nerves of wild-type mice at P21; their relative abundance is given as percent with relative standard deviation (% +/- RSD). Note that known myelin proteins constitute >80% of the total myelin protein; proteins not previously associated with myelin constitute <20%. Mass spectrometric quantification based on 3 biological replicates per genotype with 4 technical replicates each (see Figure 1—source data 1).

large proteins into gels. The present MSE-based quantification of myelin proteins also extends beyond and partially adjusts an earlier mass spectrometric approach (Patzig et al., 2011). Indeed, the current approach identified and quantified more myelin proteins, probably owing to improved protein solubilization during sample preparation and higher dynamic range of the used mass spec- trometer. By MSE, known myelin proteins (Table 1) collectively constitute over 85% of the total mye- lin protein (Figure 2) while proteins not yet associated with myelin account for the remaining 15% of the total myelin protein.

Comprehensive compendium and comparison to the transcriptome To systematically elucidate the developmental abundance profiles of the transcripts that encode peripheral myelin proteins (Figure 3), we used our combined proteome inventory of peripheral mye- lin (Figure 1—source data 1) to filter mRNA abundance data of all genes expressed in sciatic nerves. By this strategy, Figure 3 displays only those transcripts of which the protein product was identified in peripheral myelin rather than all transcripts in the nerve, thereby discriminating myelin- related mRNAs from other mRNAs such as those present in peripheral axons, fibroblasts, immune cells etc. In this assessment we additionally included PMP22 although it was not detected by MS as well as 45 proteins exclusively identified by LC-MS of myelin separated by SDS-PAGE (Figure 1— source data 1). For mRNA abundance profiles, we exploited a recently established RNA sequencing analysis (RNA-Seq; platform Illumina HiSeq 2000) of sciatic nerves dissected form wild type Sprague Dawley rats at embryonic day 21 (E21), P6, P18 and 6 months (Fledrich et al., 2018). RNA-Seq pro- vides reliable information about the relative abundance of all significantly expressed genes and is thus not limited to those represented on the previously used oligonucleotide microarrays (Patzig et al., 2011). The raw data (accessible under GEO accession number GSE115930) were nor- malized (Figure 3—source data 1) and standardized. When comparing the proteome and transcrip- tome datasets, significant mRNA abundance was detected for all 1046 transcripts for which an unambiguous unique gene identifier was found (Figure 3). 126 transcripts displayed developmen- tally unchanged abundance levels, that is, abundance changes below a threshold of 10% coefficient of variation (Figure 3B; Figure 3—source data 1).

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 6 of 31 Tools and resources Neuroscience

Table 1. Known myelin proteins in the myelin proteome. Proteins mass-spectrometrically identified in peripheral myelin are compiled according to availability of prior references as myelin pro- teins. Given are the official gene name, one selected reference, the number of transmembrane domains (TMD) and the mRNA abun- dance profile cluster (see Figure 3).

Protein name Gene Reference TMD Cluster 2-hydroxyacylsphingosine 1-beta- Ugt8 Bosio et al., 1996 2 P6-up Syntrophin a1 Snta1 Fuhrmann-Stroissnigg et al., 2012 - P18-up A2 Anxa2 Hayashi et al., 2007 - Descending Band 4.1 protein B/4.1B Epb41l3 Ivanovic et al., 2012 - Descending Band 4.1 protein G/4.1G Epb41l2 Ohno et al., 2006 - P6-up Breast carcinoma-amplified sequence 1 Bcas1 Ishimoto et al., 2017 - P6-up 1/E-Cadherin Cdh1 Fannon et al., 1995 1 P18-up Carbonic anhydrase 2 Ca2 Cammer and Tansey, 1987 - Descending a1 Ctnna1 Murata et al., 2006 - U-shaped Catenin ß1 Ctnnb1 Fannon et al., 1995 - Descending 1 Cav1 Mikol et al., 2002 1 P18-up CD9, tetraspanin 29 Cd9 Ishibashi et al., 2004 4 P18-p CD59A Cd59a Funabashi et al., 1994 1 P18-up CD47, -associated signal transducer Cd47 Gitik et al., 2011 5 P6-up CD81, tetraspanin 28 Cd81 Ishibashi et al., 2004 4 P18-up CD82, tetraspanin 27 Cd82 Chernousov et al., 2013 4 P18-up CD151, tetraspanin 24 Cd151 Patzig et al., 2011 4 P18-up molecule 4/NECL4 Cadm4 Spiegel et al., 2007 1 P6-up Cell division control protein 42 Cdc42 Benninger et al., 2007 - P6-up Cell surface MUC18 Mcam Shih et al., 1998 1 Descending Ciliary neurotrophic factor Cntf Rende et al., 1992 - Late-up CKLF-like MARVEL TMD-containing 5 Cmtm5 Patzig et al., 2011 4 P6-up -19 Cldn19 Miyamoto et al., 2005 4 P6-up Cofilin 1 Cfl1 Sparrow et al., 2012 - Descending a2 Cryab D’Antonio et al., 2006 - P18-up Cyclic nucleotide phosphodiesterase Cnp Matthieu et al., 1980 - P6-up Sarcoglycan d Sgcd Cai et al., 2007 1 Late-up Dihydropyrimidinase related protein 1 Crmp1 D’Antonio et al., 2006 - Descending Disks large homolog 1 Dlg1 Cotter et al., 2010 - Descending light chain 1 Dynll1 Myllykoski et al., 2018 - P6-up Dystroglycan Dag1 Yamada et al., 1994 1 P6-up /DP116 Dmd Cai et al., 2007 - P6-up Dystrophin-related protein 2 Drp2 Sherman et al., 2001 - P18-up E3 -protein NEDD4 Nedd4 Liu et al., 2009 - Descending Ezrin Ezr Scherer et al., 2001 - P6-up Fatty acid synthase Fasn Salles et al., 2002 - P6-up Flotillin 1 Flot1 Lee et al., 2014 - P18-up ß1 protein/Cx32 Gjb1 Li et al., 2002 4 P18-up Gap junction g3 protein/Cx29 Gjc3 Li et al., 2002 1 P6-up Gsn Gonc¸alves et al., 2010 - Late-up kinase 3ß Gsk3b Ogata et al., 2004 - P6-up Table 1 continued on next page

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 7 of 31 Tools and resources Neuroscience Table 1 continued

Protein name Gene Reference TMD Cluster Integrin a6 Itga6 Nodari et al., 2008 1 P6-up Integrin aV Itgav Chernousov and Carey, 2003 1 Descending Integrin ß1 Itgb1 Feltri et al., 2002 1 Descending Integrin ß4 Itgb4 Quattrini et al., 1996 2 P18-up Junctional adhesion molecule C Jam3 Scheiermann et al., 2007 1 P18-up Laminin a2 Lama2 Yang et al., 2005 - P6-up Laminin a4 Lama4 Yang et al., 2005 - Descending Laminin ß1 Lamb1 LeBeau et al., 1994 - Descending Laminin ß2 Lamb2 LeBeau et al., 1994 - P18-up Laminin g1 Lamc1 Chen and Strickland, 2003 - Descending Membrane Palmitoylated Protein 6 Mpp6 Saitoh et al., 2019 - P6-up -associated protein 1A Map1a Fuhrmann-Stroissnigg et al., 2012 - P18-up Microtubule-associated protein 1B Map1b Fuhrmann-Stroissnigg et al., 2012 - P6-up Mitogen-activated 1/ERK2 Mapk1 Mantuano et al., 2015 - Descending Mitogen-activated protein kinase 3/ERK1 Mapk3 Mantuano et al., 2015 - P18-up Moesin Msn Scherer et al., 2001 - Unchanged Monocarboxylate transporter 1 Slc16a1 Dome`nech-Este´vez et al., 2015 11 P18-up Myelin associated glycoprotein Mag Figlewicz et al., 1981 1 P6-up Myelin basic protein Mbp Boggs, 2006 - P6-up Myelin protein 2 Pmp2 Trapp et al., 1984 - P18-up Myelin protein zero/P0 Mpz Giese et al., 1992 1 P6-up Myelin proteolipid protein Plp1 Garbern et al., 1997 4 P6-up Myotubularin-related protein 2 Mtmr2 Bolino et al., 2004 - P6-up Noncompact myelin-associated protein Ncmap Ryu et al., 2008 1 P18-up NDRG1, N- downstream regulated Ndrg1 Berger et al., 2004 - P18-uP Neurofascin Nfasc Tait et al., 2000 2 P18-up Nidogen 1 Nid1 Lee et al., 2007 - Descending P2X purinoceptor 7 P2rÂ7 Faroni et al., 2014 - P6-up Pxn Fernandez-Valle et al., 2002 - P6-up Periaxin Prx Gillespie et al., 1994 - P6-up Plasmolipin Pllp Bosse et al., 2003 4 P18-up 1 Pfn1 Montani et al., 2014 - Descending Lin-7 homolog C Lin7c Saitoh et al., 2017 - P6-up Rac1 Rac1 Benninger et al., 2007 - U-Shaped Radixin Rdx Scherer et al., 2001 - Descending RhoA Rhoa Brancolini et al., 1999 - U-Shaped Septin 2 Sept2 Buser et al., 2009 - Descending Septin 7 Sept7 Buser et al., 2009 - U-Shaped Septin 8 Sept8 Patzig et al., 2011 - P18-up Septin 9 Sept9 Patzig et al., 2011 - P6-up Septin 11 Sept11 Buser et al., 2009 - Descending 2, NAD-dependent deacetylase Sirt2 Werner et al., 2007 - P18-up alpha chain, non-erythrocytic 1 Sptan1 Susuki et al., 2018 - P18-up Spectrin beta chain, non-erythrocytic 1 Sptbn1 Susuki et al., 2018 - P18-up Table 1 continued on next page

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 8 of 31 Tools and resources Neuroscience Table 1 continued

Protein name Gene Reference TMD Cluster protein ZO-1 Tjp1 Poliak et al., 2002 - P6-up Tight junction protein ZO-2 Tjp2 Poliak et al., 2002 - P6-up Transferrin Tf Lin et al., 1990 2 Late-up Vim Triolo et al., 2012 - Unchanged Vcl Beppu et al., 2015 - Descending

By fuzzy c-means clustering, those 920 transcripts that showed developmental abundance changes were grouped into 5 clusters (Figure 3A; Figure 3—source data 1). Among those, one cluster corresponds to an mRNA-abundance peak coinciding with an early phase of myelin biogene- sis (cluster ‘P6-UP‘), which includes the highest proportion of known myelin proteins (Table 1) such as MPZ/P0, MBP, PRX, cyclic nucleotide phosphodiesterase (CNP), fatty acid synthase (FASN), mye- lin-associated glycoprotein (MAG), proteolipid protein (PLP/DM20), -4 (CADM4/NECL4), -29 (GJC3), claudin-19 (CLDN19) and CKLF-like MARVEL-transmembrane domain containing protein-5 (CMTM5). However, many known myelin proteins clustered together according to their mRNA-abundance peak coinciding with a later phase of myelination (cluster ‘P18- UP‘), including peripheral myelin protein 2 (PMP2), tetraspanin-29 (CD9), tetraspanin-28 (CD81), con- nexin-32 (GJB1), plasmolipin (PLLP), junctional adhesion molecule-3 (JAM3), CD59 and dystrophin- related protein-2 (DRP2). The proportion of known myelin proteins was lower in the clusters corre- sponding to mRNA-abundance peaks in adulthood (clusters ‘late-UP‘, ‘U-shaped‘). Yet, a consider- able number of transcripts displayed abundance peaks at the embryonic time-point (cluster ‘Descending‘), including carbonic anhydrase 2 (CA2), cofilin-1 CFL1), beta-4 (TUBB4b) and band 4.1-protein B (EPB41L3). Generalized, the clusters were roughly similar when comparing previ- ous oligonucleotide microarray analysis of mouse sciatic nerves (Patzig et al., 2011) and the RNA- Seq analysis of rat sciatic nerves (this study); yet, the latter provides information on a larger number of genes and with a higher level of confidence. Together, clustering of mRNA abundance profiles allows categorizing peripheral myelin proteins into developmentally co-regulated groups. When systematically assessing the proteins identified in myelin by gene ontology (GO)-term anal- ysis, the functional categories over-represented in the entire myelin proteome included cell adhe- sion, and extracellular matrix (labeled in turquoise in Figure 4). When analyzing the clusters of developmentally co-expressed transcripts (from Figure 3), proteins associated with the lipid metabolism were particularly enriched in the P6-UP and P18-UP clusters, while those associated with the extracellular matrix (ECM) were over-represented in the U-shaped and Descending clusters (Figure 4). For comparison, known myelin proteins (Table 1) were over-represented in the P6-UP and P18-UP clusters (Figure 4). Together, our proteome dataset provides comprehensive in-depth coverage of the protein constituents of peripheral myelin purified from the sciatic nerves of wild type mice, and comparison to the transcriptome allows identifying developmentally co-regulated and functional groups of myelin proteins. Our data thus supply a solid resource for the molecular characterization of myelin and for discovering functionally relevant myelin proteins.

Neuropathy genes encoding myelin proteins Heritable neuropathies can be caused by mutations affecting genes preferentially expressed in neu- rons, Schwann cells or both (Rossor et al., 2013; Pareyson and Marchesi, 2009; Baets et al., 2014; Brennan et al., 2015). To systematically assess which neuropathy-causing genes encode peripheral myelin proteins, we compared our myelin proteome dataset with a current overview about disease genes at the NIH National Library of Medicine at https://ghr.nlm.nih.gov/condition/charcot-marie- tooth-disease#genes. Indeed, 31 myelin proteins were identified to be encoded by a proven neurop- athy gene (Table 2), a considerable increase compared to eight disease genes found in a similar pre- vious approach (Patzig et al., 2011). Notably, this increase is owing to both the larger size of the current myelin proteome dataset (Figure 1E) and the recent discovery of numerous neuropathy genes by the widespread application of next generation sequencing.

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 9 of 31 Tools and resources Neuroscience

A B P6-up Cluster #mRNA Aacs Cd47 Cpne3 Eml1 'ŵĩ Lama2 Mpp6 Osbpl1a Pygb Tjp1 1 P6-up 132 Abcd3 Cdc42 Cpt1a Epb41l2 Gna11 Letm1 Mpz P2rx7 Rab8b Tjp2 ϭ͘ϱ 2 P18-up 203 Acaca ŇϮ Csnk2a1 Ezr Gnai1 Lin7c Mtmr2 Pdhb Rcn1 Tspan15 3 Acat2 Ckb Cyp51 Fam177a1 Gnaq'ŵĩ Lss Mvk Pgls Rer1 Tubb2a Late-up 186 Adam10 Cldn19 Dag1 Far1 Gng2 M6pr Myh14 Plg Rpl13 Ubl3 4 U-shaped 154 Agps Cmtm5ŇϮ Dhcr7 Fasn Gsk3b'ŵĩ Mag Myo18a Plp1 Rps16 Ugt8 5 Descending 245

0 Appl1 Cnp Dhrs1 &ĚŌϭ Gypc Map1b Naga Pmp22 Rps2 ŇϮ 6 Unchanged 126 Art3 Col15a1 Dmd Fdps Hmgcs1'ŵĩ Map4 Napb Ppa2 Sept9 All 1046 Bcas1 Col1a1 Dusp15 Fmnl2 Hsd17b7 Map6 EĚƵĩϳ Prkacb Slc25a10 &ĚŌϭ Bdh1 Col1a2ŇϮ Dync1h1 Fscn1 Idi1'ŵĩ Mapre3 Ndufs8 Prx Slc25a5 Brox Col28a1 Dynll1 Fut8 Itga6 Mbp EĞŇ Psmb6 Slc44a1 &ĚŌϭ EĚƵĩϳ Cab39l Col4a2ŇϮ Eci1 Gamt Itpr3 Mical1 Nefm Psmc3 Snap91 mRNA-abundance (normalized) mRNA-abundance Cadm4 Col6a1 Ehd3 Gapdhs Kif5b Mlec Nsdhl Pura Sorbs1 оϭ͘ϱ EĞŇEĚƵĩϳ E21 P6 P18 M6 Camk2b Col6a2 Elovl1 Gjc3&ĚŌϭ Klc2 Mme Opa1 Pxn Stk39 EĞŇ &ĚŌϭ EĚƵĩϳ P18-up ^ŌϮĚϮ Abat Aldh9a1 Atp6v1e1 Cd59 Cryab Fkbp1a EĞŇHibchEĚƵĩϳ Lamb2 Mpc1 Ndufs4 Pmp2 Rab5b Serpinf1 Steap3 Tst Abhd12 Anxa11 Atp6v1h Cd81 Ctsd Flnb Hk1 Lamp1 Mpp2 Ndufs7 Por Rab7a Stx12 Tubb4a

ϭ͘ϱ ^ŌϮĚϮ Acaa1a Anxa5 Auh Cd82 Cyb5a Flot1 HrasEĞŇ Lamp2 Mpst Ndufv2 Prdx5 Rdh11 Sfxn3 Stxbp1 Vat1l Acaa2 Arl3 Bcap31 Cd9 Cyb5r3 Gatm Hsd17b10 Lancl1 Myh9 Nes Prelp Reep5 Sirt2^ŌϮĚϮ Sucla2 Vps26a Acadm Arl8b Bgn Cdh1 Ddah1 Gdi1 Hsd17b11 Ldhb Myo1b KƐƞϭNfasc Prkca Rida Slc16a1 Sult1a1 Vps29 Acads Asah1 Bphl Cdipt Decr1 Gjb1 Hsd17b12 Limch1 Myo1c Nptn Psmf1 Rtkn Slc25a1 Susd2 Vsnl1 ^ŌϮĚϮ

0 Acadvl Asl Bsg Cers2 Dnm1 Glod4 Hsd17b4 Maoa Ncdn KƐƞϭ Rab11b Rtn4 Slc2a1 Svip Ywhaq Acsl1 Atp1a1 Cadm3 Chchd3 Dpysl2 Glul Hspa12a Map1a Ncmap WŅŵPcolce Rab12 Rufy3 Slc3a2 Sypl1 ^ŌϮĚϮ Acss2 Atp1a2 Canx Clip2 Drp2 Gnao1 Hspa1a Map1lc3a Ndrg1 KƐƞϭPdcd6ip Rab21 S100a6 Slc44a2 Taldo1 ƞď Ak3 Atp1a4 Capn1 Cmtm6 Ech1 Gpi Hspa2 Map1lc3b Ndrg2 WŅŵ Rab2a Sacm1l Snta1 Tecr Aldh1a1 Atp1b3 Capn2 Cndp2 Eno2 Hadh Itgb4 Mapk3 Ndrg4 Phb Rab2b Sbds Sod2 Tkt EĚƵĩϰ KƐƞϭ Aldh1a7 Atp6v0c Cav1 Comt ƞď Hagh Jam3 Marc2 Ndufa13 WŅŵPlec Rab3a Scarb2 Sparc Tln1 mRNA-abundance (normalized) mRNA-abundance Aldh3a2 Atp6v1c1 Cavin3 Copg2 Fam213a Hepacam Jup Mgll Ndufa7 Plekhb1 Rab43 Scp2 Sptan1 Tns3 оϭ͘ϱ Ĭ KƐƞϭ E21 P6 P18 M6 Aldh6a1 Atp6v1d Cd151 Crip2 Fis1ƞď Hibadh Kit Mgst3 EĚƵĩϰ Pllp Rab4b Sept8 Sptbn1 Tom1l2 ƞĂ WŅŵ Ĭ EĚƵĩϰ ƞď WŅƉWŅŵ Late-up ƞĂ Ŷƞ Actn3 Arpc4 Cab39 Ĭ Des Esyt1 Got2 Idh3g Myl6 Pdha1 Rab18 Rras Stx7 Uqcrb ƞď EĚƵĩϭϬEĚƵĩϰ Ahcy Aspa Camk2g Cisd1 Dnajb4 ƞĂ Gpd1 Ivd Myl9 WŅƉPdlim5 Rab5c Rras2 Tagln Vat1 ϭ͘ϱ EĚƵĩϯ Akr1b1 Atp12a Capg ŶƞCkm Dnpep F13a1 Gpd1l Lap3 Ndufa5 Pebp1 Rhob Rtn1 Tagln3 Vwa5a Ĭ EĚƵĩϲEĚƵĩϰ Aldh2 Atp1a3 Capns1 Ckmt1 Dstn Fabp4 Gpx3 Lcp1 EĚƵĩϭϬNdufa6 WŅƉ Rhog S100b Tf Zc2hc1a ƞĂ Aldoa Atp1b1 Capzb Ŷƞ Dusp3 Fam129b Gpx4 Lypla2 EĚƵĩϯNdufa8 Pgam2 Rpl12 Sbspon Tomm34 Ĭ Aldoc Atp2b2 Car3 Cntn1 Dynll2 Fbxo2 Gsn Lyz2 EĚƵĩϲEĚƵĩϭϬ Picalm Rpl13a Sdha Tpi1 Anpep Atp5f1d Cast Coro1a Echs1 FggƞĂ Gstk1 Macrod1 WŅƉPkm Rpl19 Serpinb6a Tpm1 0 EĚƵĩϯ Ŷƞ Anxa1 Atp5mg Cat Cox4i1 Eef1a2 Flot2 Gstm1 Mapt EĚƵĩϲ Plin1 Rpl26 Serping1 Tppp3 Anxa3 Atp6v1g1 Cav2 Cox6c Ehd2 Fn3krp Gstm2 Mdh1 Ndufc2EĚƵĩϭϬ Plin3WŅƉ Rpl29 Sfn Tsg101 Anxa7 Bag3 ĩCavin1 CpŶƞ Ehd4 G6pd Gstm3 Me1 Npc2EĚƵĩϯ Pls3 Rpl30 Sgcd Tuba1c Aoc3 Bag5 Cavin2 Csrp1 Eif4a2 Gfap Gstp1 Mrc1 OgnEĚƵĩϲEĚƵĩϭϬ Prnp Rpl36 Slc25a20 Tuba8 EĚƵĩϯ Apoe Bcam ĩCd36 Cst3 Eno3 Glud1 Hadhb Myh11 Pacsin2 Prph Rps14 Sncg Twf2 mRNA-abundance (normalized) mRNA-abundance Apoh Blvrb Ces1d Dcn Entpd2 Gnai2 Hspa1l Myh4 PcEĚƵĩϲ Psmc6 Rps15 Sord Uchl1 оϭ͘ϱ E21 P6 P18 M6 Arl6ip5 C3 ĩ Dctn2 Ephx1 Gng3 Hspb1 Myl12b Pcmt1 Pygm Rps25 Sri Ugp2

U-shaped ĩ

Acadl Atp5f1b Cycsĩ Gclm Mat2a Ppia Rack1 Rpl23 Rpl9 Rps8 Tomm70

ϭ͘ϱ Acot7 Atp5mf Dnm3 Gnb1 Mcts1 Ppid Ralb Rpl23a Rpn2 Rpsa Tpm3 Actb Avil Dpysl4 Gng12 Ndufa12 Ppp1cb Rap1a Rpl24 Rps11 S100a10 Tuba4a Actn1 Cacybp ƞϭDsp Gpx1 Ndufa3 Ppp2r1b Rap1b Rpl27 Rps15a Sept7 Tubb3 Actn4 Calm1 Dync1i2 Hpcal1 Ndufa4 Ppp2r2a Rhoa Rpl27a Rps20 Skp1 Ube2d1 Adsl Camk2d Eef1a1 Hsp90b1 Ndufs1 Prdx1 Rhoc Rpl28 Rps23 Slc25a24 Uqcrc1

0 Ahsa1 Capza2 ƞϭEhd1 Hspa12b Nxn Prdx3 Rpl10 Rpl3 Rps24 Slc25a3 Vapa Akr1a1 Cct4 Emc1 Hspa8 Pacsin1 Prps1 Rpl11 Rpl32 Rps27a Slc25a4 Vdac3 Anp32a Cnn2 Eno1ƞϭ Ina Paics Psma6 Rpl14 Rpl35 Rps3 Snx1 Wdr1 Anxa6 Cnn3 Erp29 Kpna4 Pcbp2 Ptges3 Rpl15 Rpl4 Rps3a Snx2 Ybx3 Ap2b1 Cox6a1 ƞϭ Lasp1 Pdia4 Rab39a Rpl17 Rpl5 Rps4x Ssr4 Ywhab

mRNA-abundance (normalized) mRNA-abundance Arhgap1 Cpne6 Fbn1 Ldha Pgam1 Rab6b Rpl18 Rpl6 Rps5 Stoml2 Ywhag

оϭ͘ϱ Arpc1b Cs Fhƞϭ Lman2 Pgk1 Rab8a Rpl21 Rpl7 Rps6 Tagln2 Ywhah ^ƟƉϭ E21 P6 P18 M6 Arpc5 Ctnna1 Fhl1 Lxn Phgdh Rac1 Rpl22 Rpl7a Rps6ka3 Thy1 Zyx ^ƟƉϭ ƩŶ ^ƟƉϭ Abce1 Apex1 Calu Col18a1 Dnajb11 Fermt2 Hist1h1a Ilk Lrrc59 Nid2 Pdia3 Psmb2 Rps17 Snd1 Tomm40 Ybx1 Descending Acly Apoa1 Cand1 Copa Dnm1l Flna Hist1h1b Immt Lum Nipsnap1 Pdia6 Psmd13 Rps18 dŐĩŝSnx6 Tubb1 Ykt6 ƩŶ Actr1b Arcn1 ŇϭCapn5 Copb1 Dpysl3 Flnc Hist1h1e Ipo5 Mapk1 Nipsnap3b Pfn1 Psmd5 Rps19 ^ƟƉϭSsr1 Tubb2b Ywhae

ϭ͘ϱ Actr2 Arf4 Capza1 Cope Dpysl5 Fn1 Hist1h3a Iqgap1 Mapre1 Nme1 Phb2 Psmd7 Rrbp1 Ssr3 Tubb4b Ywhaz ƩŶ dŐĩŝ Add3 Arf5 Car2 Copg1 Dync1li1 Ganab Hist1h3b Islr Marcks Nme2 Plcd1 Ptpa Sars ^ƟƉϭ Tubb5 Zc3hav1l Ňϭ Agk Arf6 Cct2 Coro1c Eef1d Gars Hmgb1 Isoc1 EĞĬMcam Nnt Postn Sec22b Stx1b Tubb6 dŐĩŝ Aifm1 Aspn Cct3 ƩŶCrmp1 Eef1g Glg1 Hnrnpa2b1 Itgav Mthfd1 Npm1 Ppib Rap2a Sec23a Stxbp6 Tufm Ňϭ Ak2 Atp2a2 Cct5 Ctnnb1 Eif2s1 Glipr2 Hnrnpa3 Itgb1 Myadm Nras Ppic Rap2b Sec31a Surf4 Txndc5 EĞĬ 0 Akap12 Atp4a Cct6a ƩŶ Eif3b Glrx3 Hnrnpd Kif5a Myh10 Nsf Ppp1ca Rars Sec61a1 dŐĩŝSyncrip Uba1 Alad Atp5f1a ŇϭCct7 Dad1 Eif4a1 Gnai3 Hnrnpk Kif5c Nap1l1 Ola1 Ppp1cc Rcn3 Sec61b Tcp1 Ube2i EĞĬ Alcam Basp1 Cct8 Ddost Eif4h Gnas Hsp90aa1 Kpnb1 Nap1l4 Otub1 Prdx2 Rdx Sept2 dŐĩŝ Ube2n Aldh7a1 Bax Ňϭ Ddx1 Eif5a Gnb2 Hsp90ab1 Kras Ncam1 P4hb Prdx4 Rpl10a Sept11 Tln2 Usp5 Anp32b Bin1 Ckap4 Dlg1 Epb41l3 Gnb4 Hspa5 Lama4 EĞĬNcl Pa2g4 Prkaca Rpl31 Sept6 Tm9sf3 Vcl

mRNA-abundance (normalized) mRNA-abundance Anxa2 Bzw1 Ckap5 Dlst Eprs Gpd2 Hspd1 Lamb1 Nedd4 Pabpc1 Psma1 Rpl8 Serpinh1 Tmed2 Vcp

оϭ͘ϱ Ap2a2 C1qbp Cnrip1 Dnaja1 Fabp5 Gpx7 Idh2 Lamc1 EĞĬ Pafah1b2 Psma5 Rpn1 Set Tmed9 Vdac1 E21 P6 P18 M6 Ap2s1 Calr Col14a1 Dnaja2 Fam129a Hdlbp Ikbip Lpp Nid1 Pcyox1 Psma7 Rps10 Slk Tmx1 Yars

Figure 3. Developmental mRNA abundance profiles of myelin-associated genes. (A) K-means clustering was performed for the mRNA profiles of those 1046 proteins in our myelin proteome inventory for which significant mRNA expression was found by RNA-Seq in the sciatic nerve of rats dissected at ages E21, P6, P18 and 6 months (M6). Note that this filtering strategy allows to selectively display the developmental abundance profiles of those transcripts that encode myelin-associated proteins rather than of all transcripts present in the nerve. Standardized mRNA abundance profiles are shown Figure 3 continued on next page

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 10 of 31 Tools and resources Neuroscience

Figure 3 continued (n = 4 biological replicates per age). Known myelin genes are displayed in red. For comparison, Pmp22 mRNA was included although the small tetraspan protein PMP22 was not mass spectrometrically identified due to its unfavorable distribution of tryptic cleavage sites. Normalized counts for all mRNAs including those displaying developmentally unchanged abundance are provided in Figure 3—source data 1.(B) Numbers of mRNAs per cluster. The online version of this article includes the following source data for figure 3: Source data 1. Normalized developmental mRNA abundance data fi sheet 1: normalized values for all individual 4 biological replicates per age fi sheet 2: normalized values for biological replicates averaged to give mean per age.

Pathological proteomic profile of peripheral myelin in a neuropathy model The results presented thus far were based on analyzing myelin of healthy wild type mice; yet we also sought to establish a straightforward method to systematically assess myelin diversity, as exemplified by alterations in a pathological situation. As a model we chose mice carrying a homozygous deletion of the periaxin gene (Prx-/-)(Court et al., 2004; Gillespie et al., 2000). Periaxin (PRX) is the third- most abundant peripheral myelin protein (Figure 2) and scaffolds the dystroglycan complex in Schwann cells. Prx-/- mice represent an established model of Charcot-Marie-Tooth disease type 4F (Guilbot et al., 2001; Berger et al., 2006; Marchesi et al., 2010). Aiming to assess the myelin pro- teome, we purified myelin from pools of sciatic nerves dissected from Prx-/- and control mice at P21. Upon SDS-PAGE separation and silver staining the band patterns appeared roughly similar (Figure 5A), with the most obvious exception of the absence of the high-molecular weight band constituted by periaxin in Prx-/- myelin. Yet, several other bands also displayed genotype-dependent differences in intensity. As expected, PRX was also undetectable by MSE in Prx-/- myelin, in which

n.o. n.o. Protein translaƟon n.o.

n.o. Protein transport n.o.

Cytoskeleton

Extracellular matrix P6-up

n.o. P18-up n.o. Glucose metabolism n.o. n.o. Late-Up U-shaped

n.o. Lipid metabolism n.o. Descending n.o. Unchanged

Cell adhesion All proteins

Known myelin protein

0 10 20 30 [%]

Figure 4. Categorization of annotated protein functions. All proteins identified in peripheral myelin by UDMSE (turquoise) and the respective developmental expression clusters (Figure 3; shades of red) were analyzed for overrepresented functional annotations using gene ontology (GO) terms. The graph displays the percentage of proteins in each cluster that were annotated with a particular function. For comparison, known myelin proteins were annotated. n.o., not over-represented.

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 11 of 31 Tools and resources Neuroscience

Table 2. Peripheral myelin proteins identified in PNS myelin involved in neuropathological diseases. Proteins mass-spectrometically identified in peripheral myelin were analyzed regarding the involvement of the ortholog gene in neuropathological diseases. PMP22 was added, though it was not identified by MS analyses due to its unfavorable distribution of tryptic cleavage sites. CMT, Charcot-Marie-Tooth disease; DHMN, distal hereditary motor neuropathy; DI-CMTC, dominant intermedi- ate CMTC; DFN, X-linked deafness; HMN, hereditary motor neuropathy; HSAN, hereditary sensory and autonomic neuropathy; HNA, hereditary sensory and autonomic neuropathy; OMIM, Online Mendelian Inheritance in Man; PHARC, polyneuropathy, hearing loss, ataxia, and cataract; SCA, ; SPG, spastic paraplegia.

Protein name Gene name OMIM# Gene Neuropathy Monoacylglycerol lipase ABHD12 ABHD12 613599 20p11.21 Pharc -inducing factor 1 AIFM1 300169 Xq26.1 CMTX4, DFNX5 Na+/K+ -transporting ATPase a1 ATP1A1 182310 1p13.1 CMT2DD Cytochrome c oxidase subunit 6A1 COX6A1 602072 12q24.31 CMTRID Dystrophin-related protein 2 DRP2 300052 Xq22.1 CMTX subunit 1 DCTN1 601143 2p13.1 DHMN7B 2 DNM2 602378 19p13.2 CMT2M, CMTDIB Cytoplasmic dynein 1 heavy chain 1 DYNC1H1 600112 14q32.31 CMT20, SMALED1 E3 SUMO-protein ligase EGR2 129010 10q21.3 CMT1D, CMT3, CMT4E -tRNA ligase GARS (Gart) 600287 7p14.3 CMT2D, HMN5A Gap junction ß1 protein/Cx32 GJB1 304040 Xq13.1 CMTX1 Guanine nucleotide-binding protein ß4 GNB4 610863 3q26.33 CMTDIF Histidine triad nucleotide-binding protein 1 HINT1 601314 5q23.3 NMAN Hexokinase 1 HK1 142600 10q22.1 CMT4G ß1 HSPB1 602195 7q11.23 CMT2F, DHMN2B heavy chain isoform 5A KIF5A 602821 12q13.3 SPG10 Prelamin A/C LMNA 150330 1q22 CMT2B1 MME 120520 3q25.2 CMT2T, SCA43 Myelin protein zero/P0 MPZ 159440 1q23.3 CHN2,CMT1B, CMT2I, CMT2J,CMT3, CMTDID, Roussy-Levy syndrome Myotubularin-related protein 2 MTMR2 603557 11q21 CMT4B1 Alpha-N-acetylglucosaminidase NAGLU (NAGA) 609701 17q21.2 CMT2V NDRG1, N-myc downstream NDRG1 605262 8q24.22 CMT4D regulated heavy polypeptide NEFH 162230 22q12.2 CMT2CC Neurofilament light polypeptide NEFL 162280 8p21.2 CMT2E, CMT1F, CMTDIG Peripheral myelin protein 2 PMP2 170715 8q21.13 CMT1G Peripheral myelin protein 22 PMP22 601907 17p12 CMT1A, CMT1E, CMT3, HNPP, Roussy-Levy syndrome -phosphate PRPS1 311850 Xq22.3 Arts syndrome, CMTX5, DFNX1 pyrophosphokinase 1 Periaxin PRX 605725 19q13.2 CMT4F, CMT3 Ras-related protein 7a RAB7A 602298 3q21.3 CMT2B Septin 9 SEPT9 604061 17q25.3 HNA Transitional ER-ATPase VCP 601023 9p13.3 CMT2Y Tryptophan-tRNA WARS 191050 14q32.32 HMN9 ligase, cytoplasmic -tRNA YARS 603623 1p35.1 DI-CMTC ligase, cytoplasmic

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 12 of 31 Tools and resources Neuroscience

AB Protein % (+/- RSD) Protein % (+/- RSD) Myelin P0 58.13 3.75 NDRG1 0.05 0.01 -/- Ctrl Prx MBP RDX kDa 26.19 2.41 0.04 0.01 CD9 0.84 0.2 CADM4 0.04 0.00 250 PRX Others SPTBN1 0.54 0.16 SEPT2 0.03 0.00 130 TF 0.29 0.01 CMTM5 0.03 0.00 100 EPB41L2 0.28 0.05 RAC1 0.03 0.00 70 VIM 0.28 0.06 CFL1 0.02 0.00 CNP 0.25 0.03 CDC42 0.02 0.00 55 MAP1B 0.24 0.03 SEPT11 0.02 0.00 PMP2 0.23 0.02 RHOA 0.02 0.00 MBP FASN 0.20 0.02 PFN1 0.02 0.00 P0 PLP1 0.20 0.02 MPP6 0.02 0.00 35 ANXA2 0.18 0.01 SEPT7 0.02 0.00 SPTAN1 0.16 0.03 CRMP1 0.02 0.00 25 P0 PLLP 0.15 0.01 GJC3 0.02 0.00 MAG 0.11 0.01 MAPK1 0.02 0.00 CD81 0.10 0.01 EZR 0.02 0.00 MBP GSN 0.09 0.01 CAV1 0.01 0.00 CRYAB 0.06 0.01 MAPK3 0.01 0.00 MSN 0.08 0.02 DYNLL1 0.01 0.00 15 BCAS1 0.07 0.00 CNTF 0.01 0.00 CA2 0.06 0.01 GSK3B 0.01 0.00 Others 10.86 C D D‘ Prx -/- Prx -/- 15 ANXA1 15 Top 10 Immune proteins ANXA1 log FC LGALS3 AP2A1 ANXA1 2 EPB41L3 LGALS3 ATP1A4 LGALS3 4 HSD17B4 CRYAB PLCD1 HSD17B4 CORO1C CAPG 10 CNTF 10 PRX CRYAB ANXA5 CAB39L EPB41L3 CNTF 0 q-value

10 10 AHCY CAPG 10 IGH-V 5 5 IGH-3 -log LAMC1 CORO1C -log q-value CD47 IGHG2A IGKC ATP1A3 CAB39L -4 DRP2 ANXA5 n.d. MCTS1 SLC3A2 BSG PLLP 0 0 ͳlog 10 SLC16A1 PAICS q-value -2 0 2 -2 0 2 NID1 PRNP n.q. log 2 FC log 2 FC ANXA6 CNN3 18 D‘‘ ACADL LYZ2 15 ECM proteins SPTBN1 MDH1 CADM4 GJB1 E NEFM LAMP1 10 Myelin PTRF HRSP12 0

COL14A1 CSRP1 q-value -/- 10 LAMC1 Ctrl Prx CORO1A CRIP2 NID1 -log DES MYH10 5 COL14A1 COL1A2 FN1 FBN1 130 MME STXBP6 COL1A1 PCOLCE DRP2 LAMA2 ILK COL1A1 PPIC 55 COL1A2 CYB5R3 0 MCT1/ STIP1 GNG12 -2 0 2 SLC16A1 log FC HIST1H4A PDCD6IP 2 70 RALA TUBB1 D‘‘‘ BSG DUSP15 GOT2 15 Myelin proteins ALDH7A1 GATM EPB41L3 25 TUBB3 HVM51 PMP2 CRYAB DLST LYPLA2 10 MAP1A SBDS PLLP 180 PRX

ATP1A2 MYH9 q-value 10 10 DRP2 NEFL VWA5A 25 YWHAG LAP3 -log 5 SLC16A1 PLP MAG PMP2 PLP MAG FGG P0 DM20 GDI2 BASP1 MBP FBN1 RHOA 0 TUBB4 CAT -2 0 2 ATP1A1 100 NEFH CTSD log 2 FC F Ctrl Prx -/-

MAG MCT1/ SLC16a1 MAG MCT1/ SLC16a1 abaxonal adaxonal abaxonal SC Nucleus SLI adaxonal SC Nucleus * * SLI

MAG MCT1/SLC16A1 MAG MCT1/SLC16A1 * * * *

Figure 5. Molecular analysis of myelin in the Prx-/- mouse model of CMT4F. (A) Myelin purified from sciatic nerves dissected from Prx-/- and control mice at P21 was separated by SDS-PAGE (0.5 mg protein load) and proteins were visualized by silver staining. Bands constituted by the most abundant myelin proteins (MPZ/P0, MBP, PRX) are annotated. Note that no band constituted by PRX was detected in Prx-/- myelin and that several other bands also display genotype-dependent differences in intensity. Gel shows n = 2 biological replicates representative of n = 3 biological replicates. (B) The Figure 5 continued on next page

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 13 of 31 Tools and resources Neuroscience

Figure 5 continued relative abundance of proteins in myelin purified from Prx-/- sciatic nerves as quantified by MSE is given as percent with relative standard deviation (% +/- RSD). Note the increased relative abundance of MPZ/P0 and MBP compared to wild-type myelin (see Figure 2) when PRX is lacking. Mass spectrometric quantification based on 3 biological replicates with 4 technical replicates each (see Figure 5—source data 1). (C,D) Differential proteome analysis by DRE-UDMSE of myelin purified from Prx-/- and wild-type mice. Mass spectrometric quantification based on 3 biological replicates per genotype with 4 technical replicates each (see Figure 5—source data 2). (C) Top 40 proteins of which the abundance is reduced (blue) or -/- increased (red) in peripheral myelin purified from Prx compared to wild-type mice with the highest level of significance according to the -log10 transformed q-value (green). In the heatmaps, each horizontal line corresponds to the fold-change (FC) of a distinct protein compared to its average abundance in wild-type myelin plotted on a log2 color scale. Heatmaps display 12 replicates, that is 3 biological replicates per genotype with 4 technical replicates each. (D-D‘‘‘) Volcano plots representing genotype-dependent quantitative myelin proteome analysis. Data points represent quantified proteins in Prx-/- compared to wild-type myelin and are plotted as the log2-transformed fold-change (FC) on the x-axis against the -log10- transformed q-value on the y-axis. Stippled lines mark a -log10-transformed q-value of 1.301, reflecting a q-value of 0.05 as significance threshold. Highlighted are the datapoints representing the Top 10 proteins displaying highest zdist values (Euclidean distance between the two points (0,0) and (x,y) with x = log2(FC) and y = -log10(q-value) (red circles in D), immune-related proteins (purple circles in D‘), proteins of the extracellular matrix (ECM; yellow circles in D‘‘) and known myelin proteins (blue circles in D‘‘‘). n.d., not detected; n.q., no q-value computable due to protein identification in one genotype only. Also see Figure 5—figure supplement 1.(E) Immunoblot of myelin purified from Prx-/- and control sciatic nerves confirms the reduced abundance of DRP2, SLC16A1/MCT1, BSG and PMP2 in Prx-/- myelin, as found by differential DRE-UDMSE analysis (in C,D). PRX was detected as genotype control; PLP/DM20 and ATP1A1 serve as markers. Blot shows n = 2 biological replicates per genotype. (F) Teased fiber preparations of sciatic nerves dissected from Prx-/- and control mice immunolabelled for MAG (red) and SLC16A1 (green). Note that SLC16A1 co-distributes with MAG in Schmidt-Lanterman incisures (SLI) in control but not in Prx-/- nerves, in accordance with the reduced abundance of SLC16A1 in Prx-/- myelin (C–E). Also note that, in Prx-/- myelin, SLI were largely undetectable by MAG immunolabeling. The online version of this article includes the following source data and figure supplement(s) for figure 5: Source data 1. Label-free quantification of proteins in PNS myelin fractions from Prx-/- mice by MSE Identification and quantification data of detected myelin-associated proteins. Source data 2. Label-free quantification of proteins in PNS myelin fractions from WT and Prx-/- mice by DRE-UDMSE Identification and quantification data of detected myelin-associated proteins by DRE-UDMSE. Figure supplement 1. Clustered heatmap of Pearson’s correlation coefficients for protein abundance comparing genotypes.

most of the total myelin protein was constituted by MPZ/P0 and MBP (Figure 5B; Figure 5—source data 1). Upon differential analysis by DRE-UDMSE (Figure 5—source data 2), multiple proteins displayed genotype-dependent differences as visualized in a heatmap displaying those 40 proteins of which the abundance was reduced or increased with the highest statistical significance in Prx-/- compared to control myelin (Figure 5C). For example, the abundance of the periaxin-associated dystrophin- related protein 2 (DRP2) was strongly reduced in Prx-/- myelin, as previously shown by immunoblot- ting (Sherman et al., 2001). Notably, the abundance of multiple other proteins was also significantly reduced in Prx-/- myelin, including the extracellular matrix protein laminin C1 (LAMC1; previously termed LAMB2), the laminin-associated protein nidogen (NID1), Ig-like cell adhesion molecules (CADM4, MAG), the desmosomal junction protein (DES), cytoskeletal and cytoskeleton-asso- ciated proteins (EPB41L3, MAP1A, CORO1A, SPTBN1, various microtubular and intermediate fila- ment monomers), the monocarboxylate transporter MCT1 (also termed SLC16A1) and the MCT1- associated (Philp et al., 2003) immunoglobulin superfamily protein (BSG, also termed CD147). On the other hand, proteins displaying the strongest abundance increase in Prx-/- myelin included immune-related proteins (LGALS3, LYZ2, CTSD), cytoskeletal and cytoskeleton-associated proteins (CAPG, CORO1C, CNN3, several heavy chain subunits), peroxisomal enzymes (CAT, HSD17B4, MDH1) and known myelin proteins (PLLP/plasmolipin, CRYAB, GJB1/CX32). For compari- son, the abundance of the marker proteolipid protein (PLP/DM20) (Patzig et al., 2016a) and the periaxin-associated integrin beta-4 (ITGB4) (Raasakka et al., 2019) in myelin was unaltered in Prx-/- myelin. Together, differential proteome analysis finds considerably more proteins and protein groups to be altered in Prx-/- myelin than previously known (Figure 5C,D–D’’’), probably reflecting the complex pathology observed in this model (Court et al., 2004; Gillespie et al., 2000). The monocarboxylate transporter MCT1/SLC16A1 expressed by myelinating oligodendrocytes (Lee et al., 2012; Fu¨nfschilling et al., 2012) and Schwann cells (Dome`nech-Este´vez et al., 2015; Morrison et al., 2015) has been proposed to supply lactate or other glucose breakdown products to axons, in which they may serve as substrate for the mitochondrial production of ATP (Morrison et al., 2013; Saab et al., 2013; Rinholm and Bergersen, 2014). In this respect it was

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 14 of 31 Tools and resources Neuroscience striking to find the abundance of MCT1 significantly reduced in peripheral myelin when PRX is lack- ing (Figure 5C), a result that we were able to confirm by immunoblotting (Figure 5E) and immuno- labeling of teased fiber preparations of sciatic nerves (Figure 5F). Notably, reduced expression of MCT1 in Slc16a1+/- mice impairs axonal integrity at least in the CNS (Lee et al., 2012; Jha et al., 2020). The reduced abundance of MCT1 thus represents an interesting novel facet of the complex pathology in Prx-/- mice. Considering that the integrity of peripheral axons may be impaired in Prx-/- mice, we assessed their quadriceps nerves. Indeed, Prx-/- mice displayed reduced axonal diameters, a progressively reduced total number of axons and a considerable number of myelin whorls lacking a visible axon (Figure 6), indicative of impaired axonal integrity (Edgar et al., 2009). Yet we note that molecular or neuropathological features other than the reduced abundance of MCT1 probably also contribute to the axonopathy in Prx-/- mice. Together, gel-free, label free proteome analysis provides a cost- and time-efficient method that provides an accurate, sensitive tool to gain systematic insight into the protein composition of healthy peripheral myelin and its alterations in pathological situations. Indeed, gel-free proteome analysis is particularly powerful and comprehensive compared to 2D-DIGE; the workflow presented here appears readily applicable to other neuropathy models, thereby promising discovery of relevant novel features of their neuropathology.

Discussion We used gel-free, label-free quantitative mass spectrometry to assess the protein composition of myelin biochemically purified from the sciatic nerves of wild-type mice, thereby establishing a straightforward and readily applicable workflow to approach the peripheral myelin proteome. The key to comprehensiveness was to combine the strengths of three data acquisition modes, that is, MSE for correct quantification of high-abundant proteins, UDMSE for deep quantitative proteome coverage including low-abundant proteins and DRE-UDMSE for differential analysis. We suggest that DRE-UDMSE provides a good compromise between dynamic range, identification rate and instru- ment run time for routine differential myelin proteome profiling as a prerequisite for a molecular understanding of myelin (patho)biology. We have also integrated the resulting compendium with RNA-Seq-based mRNA abundance profiles in peripheral nerves and neuropathy disease loci. Beyond providing the largest peripheral myelin proteome dataset thus far, the workflow is appropriate to serve as starting point for assessing relevant variations of myelin protein composition, for example, in different nerves, ages, species and in pathological conditions. The identification of numerous pathological alterations of myelin protein composition in the Prx-/- neuropathy model indicates that the method is well suited to assess such diversity. Aiming to understand nervous system function at the molecular level, multiple ‘omics‘-scale proj- ects assess the spatio-temporal expression profiles of all mRNAs and proteins in the CNS including oligodendrocytes and myelin (Zhang et al., 2014; Patzig et al., 2016b; Sharma et al., 2015; Thakurela et al., 2016; Marques et al., 2016). Yet, peripheral nerves are also essential for normal sensory and motor capabilities. Prior approaches to the molecular profiles of Schwann cells and PNS myelin thus far, however, were performed >8 years ago (Nagarajan et al., 2002; Le et al., 2005; Ryu et al., 2008; Patzig et al., 2011; Verheijen et al., 2003; Buchstaller et al., 2004; D’Antonio et al., 2006), and the techniques have considerably advanced since. For example, current gel-free, label-free mass spectrometry can simultaneously identify and quantify the vast majority of proteins in a sample, thereby providing comprehensive in depth-information. Moreover, RNA-Seq technology has overcome limitations of the previously used microarrays for characterizing mRNA abundance profiles with respect to the number of represented genes and the suitability of the oligo- nucleotide probes. The present compendium thus provides high confidence with respect to the identification of myelin proteins, their relative abundance and their developmental mRNA expression profiles. This view is supported by the finding that over 80% of the total myelin proteome is consti- tuted by approximately 50 previously known myelin proteins. We believe that the majority of the other identified proteins represent low-abundant myelin-associated constituents in line with the high efficiency of biochemical myelin purification. Doubtless, however, the myelin proteome also com- prises contaminants from other cellular sources, underscoring the need of independent validation for establishing newly identified constituents as true myelin proteins.

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 15 of 31 iue6. Figure irgah ftliiesandsm-hnscindqarcp evsdsetda ,4ad9mnh faervasporsiels fperipheral of loss progressive reveals age of months 9 and page 4 next 2, on at continued dissected 6 nerves Figure quadriceps sectioned semi-thin toluidine-stained of micrographs Siems G E C B A tal et Relative frequency [%] Relative frequency [%] 10 15 20 10 15 20 Total axons (n) 0 5 0 5 200 400 600 Quadriceps nerve 9mo

rgesv osadrdcddaeeso eihrlaosin axons peripheral of diameters reduced and loss Progressive <0.5 0

Lf 2020;9:e51406. eLife . 0.5- <1.0 <0.5 2 mo 9 mo 0.5- <1.0

1.0- <1.5 resources and Tools 1.0- <1.5

1.5- <2.0 9mo 4mo 2mo * ** *** * 2.0- <2.5 1.5- <2.0

2.5- <3.0 2.0- <2.5

3.0- <3.5 2.5- <3.0 3.5- <4.0 3.0- <3.5 4.0- <4.5

3.5- <4.0 Axonal diameter [µm] Axonal diameter [µm] 4.5- <5.0 O:https://doi.org/10.7554/eLife.51406 DOI:

5.0- <5.5 ***p= ***p= 4.0- <4.5 5.5- <6.0

2.20e 4.5- <5.0 2.20e 6.0- <6.5

6.5- <7.0 5.0- <5.5 -16 -16

7.0- <7.5 5.5- <6.0 7.5- <8.0 6.0- <6.5 8.0- <8.5 6.5- <7.0 Myelinated axons (n) 8.5- <9.0 200 400 600

9.0- <9.5 7.0- <7.5 0 9.5- <10.0 7.5- <8.0

10.0- <10.5 9mo 4mo 2mo ***** *** ** 10.5- <11.0 8.0- <8.5

11.0 <11.5 8.5- <9.0 Ctrl Prx -/- F

ie ( mice. Relative frequency [%] 10 15 20 0 5 Prx Ctrl <0.5 0.5- <1.0 4 mo A–D -/- 1.0- <1.5

eoyedpnetqatttv seseto light of assessment quantitative Genotype-dependent ) 1.5- <2.0

2.0- <2.5

2.5- <3.0 D 3.0- <3.5 Myelin whorls (n) 100 150 50

3.5- <4.0 0

4.0- <4.5 Axonal diameter [µm] 4.5- <5.0 m m 9mo 4mo 2mo ** *** ** **

5.0- <5.5 ***p=

5.5- <6.0 2.20e 6.0- <6.5

6.5- <7.0 -16

7.0- <7.5

7.5- <8.0

8.0- <8.5

8.5- <9.0

9.0- <9.5 *

9.5- <10.0 Neuroscience Prx 10.0- <10.5 Prx Ctrl 6o 31 of 16 -/- -/- Tools and resources Neuroscience

Figure 6 continued axons in Prx-/- compared to control mice. (A) Representative micrographs. Arrows point at myelinated axons; asterisk denotes an unmyelinated axon; arrowhead points at a myelin whorl lacking a recognizable axon. Scale bars, 10 mm. (B) Total number of axons per nerve that are not associated with a Remak bundle. (C) Total number of myelinated axons per nerve. (D) Total number per nerve of myelin whorls that lack a recognizable axon. Mean +/SD, n = 3–4 mice per genotype and age; *p<0.05, **p<0.01, ***p<0.001 by Student’s unpaired t-test. (E–G) Genotype-dependent assessment of myelinated axons shows a shift toward reduced axonal diameters in quadriceps nerves of Prx-/- compared to control mice at 2 months (E), 4 months (F) and 9 months (G) of age. Data are presented as frequency distribution with 0.5 mm bin width. ***, p<0.001 by two-sided Kolmogorow-Smirnow test. For precise p-values see methods section.

Do myelin proteins exist that escape identification by standard proteomic approaches? Indeed, some proteins display atypically distributed and residues, which represent the cleav- age sites of the commonly used trypsin. The tryptic digest of these proteins leads to pepti- des that are not well suited for chromatographic separation and/or mass spectrometric detection/ sequencing, as exemplified by the small hydrophobic tetraspan-transmembrane myelin proteins MAL (Schaeren-Wiemers et al., 2004) and PMP22 (Adlkofer et al., 1995). We can thus not exclude that additional proteins with atypical tryptic digest patterns exist in peripheral myelin, which would need to be addressed by the use of alternative . Moreover, potent signaling molecules including erbB receptor tyrosine kinases (Riethmacher et al., 1997; Woldeyesus et al., 1999) and G-protein coupled receptors (GPRs) (Ackerman et al., 2018; Monk et al., 2011; Monk et al., 2009) display exceptionally low abundance. Such proteins may be identified when applying less stringent identification criteria, e.g., by requiring the sequencing of only one peptide per protein. However, lower stringency would also result in identifying false-positive proteins, which we wished to avoid for the purpose of the present compendium. We note that a truly comprehensive spatio-temporally resolved myelin proteome should preferentially also include systematic information about protein isoforms and post-translational modifications, which still poses technical challenges. Mutations affecting the periaxin (PRX) gene in cause CMT type 4F (Guilbot et al., 2001; Kabzinska et al., 2006; Bara´nkova´ et al., 2008; Tokunaga et al., 2012); the neuropathology result- ing from mutations affecting periaxin has been mainly investigated in the Prx-/- mouse model. Indeed, Prx-/- mice display a progressive including axon/myelin-units with abnormal myelin thickness, demyelination, tomaculae, onion bulbs, reduced nerve conduction veloc- ity (Gillespie et al., 2000), reduced abundance and mislocalization of the periaxin-associated DRP2 (Sherman et al., 2001) and reduced internode length (Court et al., 2004). Absence of SLIs (Gillespie et al., 2000) and bands of Cajal (Court et al., 2004) imply that the non-compact myelin compartments are impaired when PRX is lacking. In the differential analysis of myelin purified from Prx-/- and control mice we find that the previously reported reduced abundance of DRP2 (Sherman et al., 2001) represents one of the strongest molecular changes in the myelin proteome when PRX is lacking. Notably, the reported morphological changes in this neuropathy model (Sherman et al., 2001; Court et al., 2004; Gillespie et al., 2000) go along with alterations affecting the abundance of multiple other myelin-associated proteins, including junctional, cytoskeletal, extra- cellular matrix and immune-related proteins as well as lipid-modifying enzymes. Thus, the neuropa- thology in Prx-/- mice at the molecular level is more complex than previously anticipated. It is striking that the abundance of the monocarboxylate transporter MCT1/SLC16A1 that may contribute to the metabolic supply of lactate from myelinating cells to axons (Beirowski et al., 2014; Dome`nech- Este´vez et al., 2015; Kim et al., 2016; Gonc¸alves et al., 2017; Stassart et al., 2018) is strongly reduced in Prx-/- myelin. Considering that MCT1 in Schwann cells mainly localizes to Schmidt Lanter- man incisures (SLI) (Dome`nech-Este´vez et al., 2015) and that SLI are largely absent from myelin when PRX is lacking (Gillespie et al., 2000), the reduced abundance of MCT1 in Prx-/- myelin may be a consequence of the impaired myelin ultrastructure. Yet, considering that SLI are part of the cytosolic channels that may represent transport routes toward Schwann cell-dependent metabolic support of myelinated axons, the diminishment of MCT1 may contribute to reduced axonal diame- ters or axonal loss in Prx-/- mice, probably in conjunction with other molecular or morphological defects. Together, the in depth-analysis of proteins altered in neuropathy models can contribute to an improved understanding of nerve pathophysiology. Compared to a previous approach (Patzig et al., 2011), the number of proven neuropathy genes of which the encoded protein is mass spectrometrically identified in peripheral myelin has increased

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 17 of 31 Tools and resources Neuroscience four-fold from eight to 32 in the present study. This reflects both that the number of proteins identi- fied in myelin has approximately doubled and that more neuropathy genes are known due to the common use of sequencing. We note that our compendium comprises not only myelin- associated proteins causing (when mutated) demyelinating CMT1 (e.g., MPZ/P0, NEFL, PMP2) or intermediate CMT4 (GDAP1, NDRG1, PRX) but also axonal CMT2 (RAB7, GARS, HSPB1). Yet, the expression of genes causative of CMT2 is not necessarily limited to , as exemplified by the classical myelin protein MPZ/P0. Indeed, a subset of MPZ-mutations causes axonal CMT2I or CMT2J (Gallardo et al., 2009; Leal et al., 2014; Tokuda et al., 2015; Duan et al., 2016; Fabrizi et al., 2018), probably reflecting impaired axonal integrity as consequence of a primarily affect- ing Schwann cells. We also note that the nuclear EGR2/KROX20 causative of demyelinating CMT1D has not been mass spectrometrically identified in myelin, reflecting that Schwann cell nuclei are effi- ciently removed during myelin purification. While morphological analysis of peripheral nerves by light and electron microscopy is routine in numerous laboratories, systematic molecular analysis has been less straightforward. Using the sciatic nerve as a model, we show that systematic assessment of the myelin proteome and the total nerve transcriptome are suited to determine comprehensive molecular profiles in healthy nerves and in myelin-related disorders. Myelin proteome analysis can thus complement transcriptome analysis in assessing development, function and pathophysiology of peripheral nerves.

Materials and methods Mouse models Prx-/- mice (Gillespie et al., 2000) were kept on c57Bl/6 background in the animal facility of the Uni- versity of Edinburgh (United Kingdom). Genotyping was by PCR on genomic DNA using the forward primers 5‘-CAGATTTGCT CTGCCCAAGT and 5‘-CGCCTTCTAT CGCCTTCTTGAC in combination with reverse primer 5‘-ATGCCCTCAC CCACTAACAG. The PCR yielded a 0.5 kb fragment for the wildtype allele and a 0.75 kb product for the mutant allele. The age of experimental animals is given in the figure legends. All animal work conformed to United Kingdom legislation (Scientific Proce- dures) Act 1986 and to the University of Edinburgh Ethical Review Committee policy; Home Office project license No. P0F4A25E9.

Myelin purification A light-weight membrane fraction enriched for myelin was purified from sciatic nerves of mice by sucrose density centrifugation and osmotic shocks as described (Patzig et al., 2011; Erwig et al., 2019a). Myelin accumulates at the interface between 0.29 and 0.85 M sucrose. Prx-/- and wild type control C57Bl/6 mice were sacrificed by cervical dislocation at postnatal day 21 (P21). For each genotype, myelin was purified as three biological replicates (n = 3); each biological replicate repre- senting a pool of 20 sciatic nerves dissected from 10 mice. Protein concentration was determined using the DC Protein Assay Kit (Bio-Rad).

Filter-aided sample preparation for proteome analysis Protein fractions corresponding to 10 mg myelin protein were dissolved and processed according to a filter-aided sample preparation (FASP) protocol essentially as previously described for synaptic protein fractions (Ambrozkiewicz et al., 2018) and as adapted to CNS myelin (Erwig et al., 2019a; Erwig et al., 2019b). Unless stated otherwise, all steps were automated on a liquid-handling work- station equipped with a vacuum manifold (Freedom EVO 150, Tecan) by using an adaptor device constructed in-house. Briefly, myelin protein samples were lysed and reduced in lysis buffer (7 M urea, 2 M thiourea, 10 mM DTT, 0.1 M Tris pH 8.5) containing 1% ASB-14 by shaking for 30 min at 37˚C. Subsequently, the sample was diluted with ~10 volumes lysis buffer containing 2% CHAPS to reduce the ASB-14 concentration and loaded on centrifugal filter units (30 kDa MWCO, Merck Milli- pore). After removal of the detergents by washing twice with wash buffer (8 M urea, 10 mM DTT, 0.1 M Tris pH 8.5), proteins were alkylated with 50 mM iodoacetamide in 8 M urea, 0.1 M Tris pH 8.5 (20 min at RT), followed by two washes with wash buffer to remove excess reagent. Buffer was exchanged by washing three times with 50 mM ammonium bicarbonate (ABC) containing 10% ace- tonitrile. After three additional washes with 50 mM ABC/10% acetonitrile, which were performed by

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 18 of 31 Tools and resources Neuroscience centrifugation to ensure quantitative removal of liquids potentially remaining underneath the ultrafil- tration membrane, proteins were digested overnight at 37˚C with 400 ng trypsin in 40 ml of the same buffer. Tryptic peptides were recovered by centrifugation followed by two additional extraction steps with 40 ml of 50 mM ABC and 40 ml of 1% trifluoroacetic acid (TFA), respectively. Aliquots of the combined flow-throughs were spiked with 10 fmol/ml of yeast enolase-1 tryptic digest standard (Waters Corporation) for quantification purposes and directly subjected to analysis by liquid chroma- tography coupled to electrospray mass spectrometry (LC-MS). A pool of all samples was injected at least before and after any sample set to monitor stability of instrument performance.

Mass spectrometry Nanoscale reversed-phase UPLC separation of tryptic peptides was performed with a nanoAcquity UPLC system equipped with a Symmetry C18 5 mm, 180 mm  20 mm trap column and a HSS T3 C18 1.8 mm, 75 mm  250 mm analytical column (Waters Corporation) maintained at 45˚C. Injected peptides were trapped for 4 min at a flow rate of 8 ml/min 0.1% TFA and then separated over 120 min at a flow rate of 300 nl/min with a gradient comprising two linear steps of 3–35% mobile phase B in 105 min and 35–60% mobile phase B in 15 min, respectively. Mobile phase A was water contain- ing 0.1% formic acid while mobile phase B was acetonitrile containing 0.1% formic acid. Mass spec- trometric analysis of tryptic peptides was performed using a Synapt G2-S quadrupole time-of-flight mass spectrometer equipped with ion mobility option (Waters Corporation). Positive ions in the mass range m/z 50 to 2000 were acquired with a typical resolution of at least 20.000 FWHM (full width at half maximum) and data were lock mass corrected post-acquisition. UDMSE and DRE- UDMSE analyses were performed in the ion mobility-enhanced data-independent acquisition mode with drift time-specific collision energies as described in detail by Distler et al. (2016) and Distler et al. (2014b). Specifically, for DRE-UDMSE a deflection device (DRE lens) localized between the quadrupole and the ion mobility cell of the mass spectrometer was cycled between full (100% for 0.4 s) and reduced (5% for 0.4 s) ion transmission during one 0.8 s full scan. Continuum LC-MS data were processed for signal detection, peak picking, and isotope and charge state deconvolution using Waters ProteinLynx Global Server (PLGS) version 3.0.2 (47). For protein identification, a custom database was compiled by adding the sequence information for yeast enolase 1 and porcine trypsin to the UniProtKB/Swiss-Prot mouse proteome and by appending the reversed sequence of each entry to enable the determination of false discovery rate (FDR). Precursor and fragment ion mass tol- erances were automatically determined by PLGS 3.0.2 and were typically below 5 ppm for precursor ions and below 10 ppm (root mean square) for fragment ions. Carbamidomethylation of was specified as fixed and oxidation of as variable modification. One missed trypsin cleavage was allowed. Minimal ion matching requirements were two fragments per peptide, five fragments per protein, and one peptide per protein. The FDR for protein identification was set to 1% threshold.

Analysis of proteomic data For each genotype (Prx-/- and wild type control mice sacrificed at P21), biochemical fractions enriched for PNS myelin were analyzed as three biological replicates (n = 3 per condition); each bio- logical replicate representing a pool of 20 sciatic nerves dissected from 10 mice. The samples were processed with replicate digestion and injection, resulting in four technical replicates per biological replicate and thus a total of 12 LC-MS runs per condition to be compared, essentially as previously reported for CNS myelin (Patzig et al., 2016b; Erwig et al., 2019b). The freely available software ISOQuant (www.isoquant.net) was used for post-identification analysis including retention time align- ment, exact mass and retention time (EMRT) and ion mobility clustering, peak intensity normaliza- tion, isoform/ filtering and calculation of absolute in-sample amounts for each detected protein (Distler et al., 2016; Distler et al., 2014b; Kuharev et al., 2015) according to the TOP3 quantification approach (Silva et al., 2006; Ahrne´ et al., 2013). Only peptides with a minimum length of seven amino acids that were identified with scores above or equal to 5.5 in at least two runs were considered. FDR for both peptides and proteins was set to 1% threshold and only proteins reported by at least two peptides (one of which unique) were quantified using the TOP3 method. The parts per million (ppm) abundance values (i.e. the relative amount (w/w) of each protein in respect to the sum over all detected proteins) were log2-transformed and normalized by subtraction

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 19 of 31 Tools and resources Neuroscience of the median derived from all data points for the given protein. Significant changes in protein abun- dance were detected by moderated t-statistics essentially as described (Ambrozkiewicz et al., 2018; Erwig et al. (2019b) ) across all technical replicates using an empirical Bayes approach and false discovery (FDR)-based correction for multiple comparisons (Kammers et al., 2015). For this purpose, the Bioconductor R packages ‘limma’ (Ritchie et al., 2015) and ‘q-value’ (Storey, 2003) were used in RStudio, an integrated development environment for the open source programming language R. Proteins identified as contaminants (e.g. components of or hair cells) were removed from the analysis. Proteins with ppm values below 100 which were not identified in one genotype were considered as just above detection level and also removed from the analysis. The rel- ative abundance of a protein in myelin was accepted as altered if both statistically significant (q- value <0.05). Pie charts, heatmaps and volcano plots were prepared in Microsoft Excel 2013 and GraphPad Prism 7. Pearson’s correlation coefficients derived from log2-transformed ppm abundance values were clustered and visualized with the tool heatmap.2 contained in the R package gplots (CRAN.R-project.org/package = gplots). Only pairwise complete observations were considered to reduce the influence of missing values on clustering behavior. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium (proteomecentral.proteomex- change.org) via the PRIDE partner repository (Vizcaı´no et al., 2016) with the dataset identifier PXD015960.

Gel electrophoresis and silver staining of gels Protein concentration was determined using the DC Protein Assay (BioRad). Samples were sepa- rated on a 12% SDS-PAGE for 1 hr at 200 V using the BioRad system, fixated overnight in 10% [v/v]

acetic acid and 40% [v/v] ethanol and then washed in 30% ethanol (2 Â 20 min) and ddH2O (1 Â 20

min). For sensitization, gels were incubated 1 min in 0.012% [v/v] Na2S2O3 and subsequently washed

with ddH2O (3 Â 20 s). For silver staining, gels were impregnated for 20 min in 0.2% [w/v] AgNO3/

0.04% formaldehyde, washed with ddH2O (3 Â 20 s) and developed in 3% [w/v] Na2CO3/0.02% [w/v] formaldehyde. The reaction was stopped by exchanging the solution with 5% [v/v] acetic acid.

Immunoblotting Immunoblotting was performed as described by Schardt et al. (2009) and de Monasterio-Schrader et al. (2013). Primary were specific for dystrophin-related-protein 2 (DRP2; Sigma; 1:1000), peripheral myelin protein 2 (PMP2; ProteinTech Group 12717–1-AP; 1:1000), proteolipid protein (PLP/DM20; A431; Jung et al., 1996; 1:5000), Monocarboxylate transporter 1 (MCT1/ SLC16A1; Stumpf et al., 2019; 1:1000), periaxin (PRX; Gillespie et al., 1994; 1:1000), sodium/potas- sium-transporting ATPase subunit alpha-1 (ATP1A1; 1:2000; Abcam #13736–1-AP), myelin protein zero (MPZ/P0; Archelos et al., 1993; kind gift by J Archelos-Garcia; 1:10.000), voltage-dependent anion-selective channel protein (VDAC; Abcam #ab15895; 1:1000), basigin (BSG/CD147; Protein- Tech Group #ab64616; 1:1000), neurofilament H (NEFH/NF-H; Covance #SMI-32P; 1:1000), voltage- gated potassium channel subunit A member 1 (KCNA1; Neuromab #73–007; 1:1000), EGR2/ KROX20 (Darbas et al., 2004; kind gift by D Meijer, Edinburgh; 1:1000) and myelin basic protein (MBP; 1:2000). To generate the latter antisera, rabbits were immunized (Pineda Antiko¨ rper Service, Berlin, Germany) with the KLH-coupled peptide CQDENPVVHFFK corresponding to amino acids 212–222 of mouse MBP isoform 1 (Swisprot/Uniprot-identifier P04370-1). Anti-MBP antisera were purified by affinity chromatography and extensively tested for specificity by immunoblot analysis of homogenate of dissected from wild-type mice compared to Mbpshiverer/shiverer mice that lack expression of MBP. Appropriate secondary anti-mouse or -rabbit antibodies conjugated to HRP were from dianova. Immunoblots were developed using the Enhanced Chemiluminescence Detec- tion kit (Western Lightning Plus, Perkin Elmer) and detected with the Intas ChemoCam system (INTAS Science Imaging Instruments GmbH, Go¨ ttingen, Germany).

Immunolabelling of teased fibers Teased fibers were prepared as previously described by Sherman et al. (2001) and Catenaccio and Court (2018). For each genotype, one male mouse was sacrificed by cervical dislocation at P17. Immunolabelling of teased fibers was performed as described by Patzig et al. (2016b). Briefly, teased fibers were fixed for 5 min in 4% paraformaldehyde, permeabilized 5 min with ice-cold

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 20 of 31 Tools and resources Neuroscience methanol, washed in PBS (3 Â 5 min) and blocked for 1 hr at 21˚C in blocking buffer (10% horse serum, 0.25% Triton X-100, 1% bovine serum albumin in PBS). Primary antibodies were applied over- night at 4˚C in incubation buffer (1.5% horse serum, 0.25% Triton X-100 in PBS). Samples were washed in PBS (3 Â 5 min) and secondary antibodies were applied in incubation buffer (1 hr, RT). Samples were again washed in PBS (2 Â 5 min), and 4‘,6-diamidino-2-phenylindole (DAPI; 1:50 000

in PBS) was applied for 10 min at RT. Samples were briefly washed 2x with ddH2O and mounted using Aqua-Poly/Mount (Polysciences, Eppelheim, Germany). Antibodies were specific for myelin- associated glycoprotein (MAG clone 513; Chemicon MAB1567; 1:50) and MCT1/SLC16A1 (107). Sec- ondary antibodies were donkey a-rabbit-Alexa488 (Invitrogen A21206; 1:1000) and donkey a- mouse-Alexa555 (Invitrogen A21202; 1:1000). Labeled teased fibers were imaged using the confocal microscope Leica SP5. The signal was collected with the objective HCX PL APO lambda blue 63.0. x1.20. DAPI staining was excited with 405 nm and collected between 417 nm - 480 nm. To excite the Alexa488 fluorophore an Argon laser with the excitation of 488 nm was used and the emission was set to 500 nm - 560 nm. Alexa555 was excited by using the DPSS561 laser at an excitation of 561 nm and the emission was set to 573 nm - 630 nm. To export and process the images LAS AF lite and Adobe Photoshop were used.

mRNA abundance profiles Raw data were previously established (Fledrich et al., 2018) from the sciatic nerves of wild type Sprague Dawley rats at the indicated ages (E21, P6, P18; n = 4 per time point). Briefly, sciatic nerves were dissected, the epineurium was removed, total RNA was extracted with the RNeasy Kit (Qia- gen), concentration and quality (ratio of absorption at 260/280 nm) of RNA samples were deter- mined using the NanoDrop spectrophotometer (ThermoScientific), integrity of the extracted RNA was determined with the Agilent 2100 Bioanalyser (Agilent Technologies) and RNA-Seq was per- formed using the Illumina HiSeq2000 platform. RNA-Seq raw data are available under the GEO accession number GSE115930 (Fledrich et al., 2018). For the present analysis, the fastqfiles were mapped to rattus norvegicus rn6 using Tophat Aligner and then quantified based on the Ensemble Transcripts release v96. The raw read counts were then normalized using the R package DESeq2. The normalized data was then standardized to a mean of zero and a standard devi- ation of one, therefore genes with similar changes in expression are close in the euclidian space. Clustering was performed on the standardized data using the R package mfuzz. Transcripts display- ing abundance differences of less than 10% coefficient of variation were considered developmentally unchanged.

Venn diagrams Area-proportional Venn diagrams were prepared using BioVenn (Hulsen et al., 2008) at www.bio- venn.nl/.

GO-term For functional categorization of the myelin proteome the associated gene ontology terms were sys- tematically analyzed on the mRNA abundance cluster using the Database for Annotation, Visualiza- tion and Integrated Discovery (DAVID; https://david.ncifcrf.gov). For comparison known myelin proteins according to literature were added.

Histological analysis Prx-/- and control mice were perfused at the indicated ages intravascularly with fixative solution (2.5% glutaraldehyde, 4% paraformaldehyde, 0.1 M sodium cacodylate buffer, pH 7.4). Quadriceps nerves were removed, fixed for 2 hr at room temperature, followed by 18 hr at 4˚C in the same fixa-

tive, postfixed in OsO4, dehydrated a graded series of ethanol, followed by propylene oxide and embedded in Araldite. All axons not associated with a Remak bundle were counted and categorized as myelinated or non-myelinated. All myelin profiles lacking a recognizable axon were counted. The total number of axons were counted on micrographs of toluidine blue stained Araldite sections (0.5 mm) of quadriceps nerves. Precise p-values for the quantitative comparison between Ctrl and Prx-/- mice were: Total number of axons (Figure 6B; Student’s unpaired t-test): 2 mo p=0.01734; 4 mo p=2.1E-05; 9 mo p=0.007625; Number of myelinated axons (Figure 6C; Student’s unpaired t-test): 2

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 21 of 31 Tools and resources Neuroscience mo p=0.00444; 4 mo p=2.12E-05; 9 mo p=0.005766; Number of empty myelin profiles (Figure 6D; Student’s unpaired t-test): 2 mo p=0.004445; 4 mo p=0.001461; 9 mo p=0.000695; Axonal diame- ters (Figure 6E–G; two-sided Kolmogorow-Smirnow test): 2 mo p=2.20E-16; 4 mo p=2.20E-16; 9 mo p=2.20E-16.

Acknowledgements We thank J Archelos-Garcia and D Meijer for antibodies, T Buscham and J Edgar for discussions, L Piepkorn for support in data analysis, K-A Nave for support made possible by a European Research Council Advanced Grant (‘MyeliNano‘ to K-AN) and the International Max Planck Research School for Genome Science (IMPRS-GS) for supporting SBS.

Additional information

Funding Funder Grant reference number Author Deutsche Forschungsge- WE 2720/2-2 Hauke B. Werner meinschaft Deutsche Forschungsge- WE 2720/4-1 Hauke B. Werner meinschaft Deutsche Forschungsge- WE 2720/5-1 Hauke B. Werner meinschaft Deutsche Forschungsge- RO 4076/3-2 Moritz J. Rossner meinschaft Wellcome 0842424 Peter Brophy

The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Author contributions Sophie B Siems, Performed all experiments not specified otherwise, conducted statistical analysis, contributed to analysis and interpretation of data; Olaf Jahn, Performed proteome analysis, contrib- uted to analysis and interpretation of data and writing the article; Maria A Eichel, Performed teased fiber labeling and microscopy; Nirmal Kannaiyan, Performed bioinformatic analysis of RNA-Seq data; Lai Man N Wu, Diane L Sherman, Performed histological analysis; Kathrin Kusch, Provided unpub- lished reagents; Do¨ rte Hesse, Contributed to proteome analysis; Ramona B Jung, Performed bio- chemical purification of myelin; Robert Fledrich, Michael W Sereda, Provided an unpublished RNA- Seq dataset; Moritz J Rossner, Supervised bioinformatic analysis of RNA-Seq data; Peter J Brophy, Supervised histological analysis; Hauke B Werner, Conceived, designed and directed the study, ana- lyzed and interpreted data and wrote the article

Author ORCIDs Olaf Jahn https://orcid.org/0000-0002-3397-8924 Kathrin Kusch https://orcid.org/0000-0002-5079-502X Hauke B Werner https://orcid.org/0000-0002-7710-5738

Ethics Animal experimentation: All animal work conformed to United Kingdom legislation (Scientific Proce- dures) Act 1986 and to the University of Edinburgh Ethical Review Committee policy; Home Office project license No. P0F4A25E9.

Decision letter and Author response Decision letter https://doi.org/10.7554/eLife.51406.sa1 Author response https://doi.org/10.7554/eLife.51406.sa2

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 22 of 31 Tools and resources Neuroscience

Additional files Supplementary files . Transparent reporting form

Data availability All data generated or analysed during this study are included in the manuscript and supporting files. This includes the mass spectrometry proteomics data. Source data files have been provided for Fig- ures 1, 3 and 5. Additional to being provided in the source data files, mass spectrometry proteomics data have been deposited to the PRIDE/ProteomeXchange Consortium with dataset identifier PXD015960.

The following dataset was generated:

Database and Author(s) Year Dataset title Dataset URL Identifier Olaf Jahn 2020 Proteome profile of peripheral https://www.ebi.ac.uk/ PRIDE, PXD015960 myelin in healthy mice and in a pride/archive/projects/ neuropathy model PXD015960

References Ackerman SD, Luo R, Poitelon Y, Mogha A, Harty BL, D’Rozario M, Sanchez NE, Lakkaraju AKK, Gamble P, Li J, Qu J, MacEwan MR, Ray WZ, Aguzzi A, Feltri ML, Piao X, Monk KR. 2018. GPR56/ADGRG1 regulates development and maintenance of peripheral myelin. Journal of Experimental Medicine 215:941–961. DOI: https://doi.org/10.1084/jem.20161714, PMID: 29367382 Adlkofer K, Martini R, Aguzzi A, Zielasek J, Toyka KV, Suter U. 1995. Hypermyelination and demyelinating peripheral neuropathy in Pmp22-deficient mice. Nature 11:274–280. DOI: https://doi.org/10.1038/ ng1195-274, PMID: 7581450 Ahrne´ E, Molzahn L, Glatter T, Schmidt A. 2013. Critical assessment of proteome-wide label-free absolute abundance estimation strategies. Proteomics 13:2567–2578. DOI: https://doi.org/10.1002/pmic.201300135 Ambrozkiewicz MC, Schwark M, Kishimoto-Suga M, Borisova E, Hori K, Salazar-La´zaro A, Rusanova A, Altas B, Piepkorn L, Bessa P, Schaub T, Zhang X, Rabe T, Ripamonti S, Rosa´rio M, Akiyama H, Jahn O, Kobayashi T, Hoshino M, Tarabykin V, et al. 2018. Polarity acquisition in cortical neurons is driven by synergistic action of Sox9-Regulated Wwp1 and Wwp2 E3 ubiquitin and intronic miR-140. 100:1097–1115. DOI: https://doi.org/10.1016/j.neuron.2018.10.008, PMID: 30392800 Archelos JJ, Roggenbuck K, Schneider-Schaulies J, Linington C, Toyka KV, Hartung HP. 1993. Production and characterization of monoclonal antibodies to the extracellular domain of P0. Journal of Neuroscience Research 35:46–53. DOI: https://doi.org/10.1002/jnr.490350107, PMID: 7685397 Baets J, De Jonghe P, Timmerman V. 2014. Recent advances in Charcot-Marie-Tooth disease. Current Opinion in Neurology 27:532–540. DOI: https://doi.org/10.1097/WCO.0000000000000131, PMID: 25110935 Bara´ nkova´ L, Siskova´D, Hu¨ hne K, Vyhna´lkova´E, Sakmaryova´I, Bojar M, Rautenstrauss B, Seeman P. 2008. A 71- nucleotide deletion in the periaxin gene in a Romani patient with early-onset slowly progressive demyelinating CMT. European Journal of Neurology 15:548–551. DOI: https://doi.org/10.1111/j.1468-1331.2008.02104.x, PMID: 18410371 Beirowski B, Babetto E, Golden JP, Chen YJ, Yang K, Gross RW, Patti GJ, Milbrandt J. 2014. Metabolic regulator LKB1 is crucial for Schwann cell-mediated axon maintenance. Nature Neuroscience 17:1351–1361. DOI: https://doi.org/10.1038/nn.3809, PMID: 25195104 Benninger Y, Thurnherr T, Pereira JA, Krause S, Wu X, Chrostek-Grashoff A, Herzog D, Nave KA, Franklin RJ, Meijer D, Brakebusch C, Suter U, Relvas JB. 2007. Essential and distinct roles for and in the regulation of Schwann cell biology during peripheral nervous system development. The Journal of Cell Biology 177:1051–1061. DOI: https://doi.org/10.1083/jcb.200610108, PMID: 17576798 Beppu M, Sawai S, Satoh M, Mori M, Kazami T, Misawa S, Shibuya K, Ishibashi M, Sogawa K, Kado S, Kodera Y, Nomura F, Kuwabara S. 2015. Autoantibodies against vinculin in patients with chronic inflammatory demyelinating polyneuropathy. Journal of Neuroimmunology 287:9–15. DOI: https://doi.org/10.1016/j. jneuroim.2015.07.012, PMID: 26439954 Berger P, Sirkowski EE, Scherer SS, Suter U. 2004. Expression analysis of the N-Myc downstream-regulated gene 1 indicates that myelinating Schwann cells are the primary disease target in hereditary motor and sensory neuropathy-Lom. Neurobiology of Disease 17:290–299. DOI: https://doi.org/10.1016/j.nbd.2004.07.014, PMID: 15474366 Berger P, Niemann A, Suter U. 2006. Schwann cells and the pathogenesis of inherited motor and sensory neuropathies (Charcot-Marie-Tooth disease). Glia 54:243–257. DOI: https://doi.org/10.1002/glia.20386, PMID: 16856148

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 23 of 31 Tools and resources Neuroscience

Boggs JM. 2006. Myelin basic protein: a multifunctional protein. Cellular and Molecular Life Sciences 63:1945– 1961. DOI: https://doi.org/10.1007/s00018-006-6094-7, PMID: 16794783 Bolino A, Bolis A, Previtali SC, Dina G, Bussini S, Dati G, Amadio S, Del Carro U, Mruk DD, Feltri ML, Cheng CY, Quattrini A, Wrabetz L. 2004. Disruption of Mtmr2 produces CMT4B1-like neuropathy with myelin outfolding and impaired . The Journal of Cell Biology 167:711–721. DOI: https://doi.org/10.1083/jcb. 200407010, PMID: 15557122 Bosio A, Binczek E, Stoffel W. 1996. Functional breakdown of the lipid bilayer of the myelin membrane in central and peripheral nervous system by disrupted galactocerebroside synthesis. PNAS 93:13280–13285. DOI: https://doi.org/10.1073/pnas.93.23.13280, PMID: 8917582 Bosse F, Hasse B, Pippirs U, Greiner-Petter R, Mu¨ ller HW. 2003. Proteolipid plasmolipin: localization in polarized cells, regulated expression and lipid raft association in CNS and PNS myelin. Journal of Neurochemistry 86: 508–518. DOI: https://doi.org/10.1046/j.1471-4159.2003.01870.x, PMID: 12871592 Brancolini C, Marzinotto S, Edomi P, Agostoni E, Fiorentini C, Mu¨ ller HW, Schneider C. 1999. Rho-dependent regulation of cell spreading by the tetraspan Gas3/PMP22. Molecular Biology of the Cell 10:2441–2459. DOI: https://doi.org/10.1091/mbc.10.7.2441, PMID: 10397775 Brennan KM, Bai Y, Shy ME. 2015. Demyelinating CMT–what’s known, what’s new and what’s in store? Neuroscience Letters 596:14–26. DOI: https://doi.org/10.1016/j.neulet.2015.01.059 Brostoff SW, Karkhanis YD, Carlo DJ, Reuter W, Eylar EH. 1975. Isolation and partial characterization of the major proteins of rabbit sciatic nerve myelin. Brain Research 86:449–458. DOI: https://doi.org/10.1016/0006- 8993(75)90895-1 Buchstaller J, Sommer L, Bodmer M, Hoffmann R, Suter U, Mantei N. 2004. Efficient isolation and gene expression profiling of small numbers of stem cells and developing Schwann cells. Journal of Neuroscience 24:2357–2365. DOI: https://doi.org/10.1523/JNEUROSCI.4083-03.2004, PMID: 15014110 Buser AM, Erne B, Werner HB, Nave KA, Schaeren-Wiemers N. 2009. The septin cytoskeleton in myelinating glia. Molecular and Cellular Neuroscience 40:156–166. DOI: https://doi.org/10.1016/j.mcn.2008.10.002, PMID: 1 9026747 Cai H, Erdman RA, Zweier L, Chen J, Shaw JH, Baylor KA, Stecker MM, Carey DJ, Chan YM. 2007. The sarcoglycan complex in Schwann cells and its role in myelin stability. Experimental Neurology 205:257–269. DOI: https://doi.org/10.1016/j.expneurol.2007.02.015, PMID: 17397833 Cammer W, Tansey FA. 1987. Immunocytochemical localization of carbonic anhydrase in myelinated fibers in peripheral nerves of rat and mouse. Journal of Histochemistry & Cytochemistry 35:865–870. DOI: https://doi. org/10.1177/35.8.3110266, PMID: 3110266 Catenaccio A, Court FA. 2018. Teased fiber preparation of myelinated nerve fibers from peripheral nerves for vital dye staining and immunofluorescence analysis. Methods in Molecular Biology 1739:329–337. DOI: https:// doi.org/10.1007/978-1-4939-7649-2_21, PMID: 29546717 Chen ZL, Strickland S. 2003. Laminin gamma1 is critical for Schwann cell differentiation, axon myelination, and regeneration in the peripheral nerve. Journal of Cell Biology 163:889–899. DOI: https://doi.org/10.1083/jcb. 200307068, PMID: 14638863 Chernousov MA, Yu WM, Chen ZL, Carey DJ, Strickland S. 2008. Regulation of Schwann cell function by the extracellular matrix. Glia 56:1498–1507. DOI: https://doi.org/10.1002/glia.20740, PMID: 18803319 Chernousov MA, Stahl RC, Carey DJ. 2013. are involved in Schwann cell-axon interaction. Journal of Neuroscience Research 91:1419–1428. DOI: https://doi.org/10.1002/jnr.23272, PMID: 24038174 Chernousov MA, Carey DJ. 2003. alphaVbeta8 integrin is a Schwann cell receptor for fibrin. Experimental Cell Research 291:514–524. DOI: https://doi.org/10.1016/S0014-4827(03)00409-9, PMID: 14644171 Cotter L, Ozc¸elik M, Jacob C, Pereira JA, Locher V, Baumann R, Relvas JB, Suter U, Tricaud N. 2010. Dlg1-PTEN interaction regulates myelin thickness to prevent damaging peripheral nerve overmyelination. Science 328: 1415–1418. DOI: https://doi.org/10.1126/science.1187735, PMID: 20448149 Court FA, Sherman DL, Pratt T, Garry EM, Ribchester RR, Cottrell DF, Fleetwood-Walker SM, Brophy PJ. 2004. Restricted growth of Schwann cells lacking Cajal bands slows conduction in myelinated nerves. Nature 431: 191–195. DOI: https://doi.org/10.1038/nature02841, PMID: 15356632 D’Antonio M, Michalovich D, Paterson M, Droggiti A, Woodhoo A, Mirsky R, Jessen KR. 2006. Gene profiling and bioinformatic analysis of Schwann cell embryonic development and myelination. Glia 53:501–515. DOI: https://doi.org/10.1002/glia.20309, PMID: 16369933 Darbas A, Jaegle M, Walbeehm E, van den Burg H, Driegen S, Broos L, Uyl M, Visser P, Grosveld F, Meijer D. 2004. Cell autonomy of the mouse claw paw mutation. Developmental Biology 272:470–482. DOI: https://doi. org/10.1016/j.ydbio.2004.05.017, PMID: 15282162 de Monasterio-Schrader P, Jahn O, Tenzer S, Wichert SP, Patzig J, Werner HB. 2012. Systematic approaches to myelin. Cellular and Molecular Life Sciences 69:2879–2894. DOI: https://doi.org/10. 1007/s00018-012-0958-9 de Monasterio-Schrader P, Patzig J, Mo¨ bius W, Barrette B, Wagner TL, Kusch K, Edgar JM, Brophy PJ, Werner HB. 2013.Uncoupling of neuroinflammation from axonal degeneration in mice lacking the myelin protein tetraspanin-2. Glia 61:1832–1847. DOI: https://doi.org/10.1002/glia.22561 Distler U, Kuharev J, Tenzer S. 2014a. Biomedical applications of ion mobility-enhanced data-independent acquisition-based label-free quantitative proteomics. Expert Review of Proteomics 11:675–684. DOI: https:// doi.org/10.1586/14789450.2014.971114, PMID: 25327648

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 24 of 31 Tools and resources Neuroscience

Distler U, Kuharev J, Navarro P, Levin Y, Schild H, Tenzer S. 2014b. Drift time-specific collision energies enable deep-coverage data-independent acquisition proteomics. Nature Methods 11:167–170. DOI: https://doi.org/ 10.1038/nmeth.2767 Distler U, Kuharev J, Navarro P, Tenzer S. 2016. Label-free quantification in ion mobility-enhanced data- independent acquisition proteomics. Nature Protocols 11:795–812. DOI: https://doi.org/10.1038/nprot.2016. 042, PMID: 27010757 Dodds JN, Baker ES. 2019. Ion mobility spectrometry: fundamental concepts, instrumentation, applications, and the road ahead. Journal of the American Society for Mass Spectrometry 30:2185–2195. DOI: https://doi.org/ 10.1007/s13361-019-02288-2, PMID: 31493234 Dome` nech-Este´ vez E, Baloui H, Repond C, Rosafio K, Me´dard JJ, Tricaud N, Pellerin L, Chrast R. 2015. Distribution of monocarboxylate transporters in the peripheral nervous system suggests putative roles in lactate shuttling and myelination. Journal of Neuroscience 35:4151–4156. DOI: https://doi.org/10.1523/ JNEUROSCI.3534-14.2015, PMID: 25762662 Duan X, Gu W, Hao Y, Wang R, Wen H, Sun S, Jiao J, Fan D. 2016. A novel Asp121Asn mutation of myelin protein zero is associated with late-onset axonal Charcot-Marie-Tooth disease, hearing loss and pupil abnormalities. Frontiers in Aging Neuroscience 8:00222. DOI: https://doi.org/10.3389/fnagi.2016.00222 Edgar JM, McLaughlin M, Werner HB, McCulloch MC, Barrie JA, Brown A, Faichney AB, Snaidero N, Nave KA, Griffiths IR. 2009. Early ultrastructural defects of axons and axon-glia junctions in mice lacking expression of Cnp1. Glia 57:1815–1824. DOI: https://doi.org/10.1002/glia.20893, PMID: 19459211 Erwig MS, Hesse D, Jung RB, Uecker M, Kusch K, Tenzer S Jahn O Werner HB. 2019a. Myelin: Methods for Purification and Proteome Analysis. In: Methods in Molecular Biology. Springer. p. 37–63. DOI: https://doi.org/ 10.1007/978-1-4939-9072-6_3 Erwig MS, Patzig J, Steyer AM, Dibaj P, Heilmann M, Heilmann I, Jung RB, Kusch K, Mo¨ bius W, Jahn O, Nave KA, Werner HB. 2019b. Anillin facilitates septin assembly to prevent pathological outfoldings of central nervous system myelin. eLife 8:e43888. DOI: https://doi.org/10.7554/eLife.43888, PMID: 30672734 Fabrizi GM, Tamburin S, Cavallaro T, Cabrini I, Ferrarini M, Taioli F, Magrinelli F, Zanette G. 2018. The spectrum of Charcot-Marie-Tooth disease due to myelin protein zero: an electrodiagnostic, nerve ultrasound and histological study. Clinical Neurophysiology 129:21–32. DOI: https://doi.org/10.1016/j.clinph.2017.09.117, PMID: 29136549 Fannon AM, Sherman DL, Ilyina-Gragerova G, Brophy PJ, Friedrich VL, Colman DR. 1995. Novel E-cadherin- mediated adhesion in peripheral nerve: Schwann cell architecture is stabilized by autotypic adherens junctions. Journal of Cell Biology 129:189–202. DOI: https://doi.org/10.1083/jcb.129.1.189, PMID: 7698985 Faroni A, Smith RJ, Procacci P, Castelnovo LF, Puccianti E, Reid AJ, Magnaghi V, Verkhratsky A. 2014. Purinergic signaling mediated by P2X7 receptors controls myelination in sciatic nerves. Journal of Neuroscience Research 92:1259–1328. DOI: https://doi.org/10.1002/jnr.23417, PMID: 24903685 Feltri ML, Graus Porta D, Previtali SC, Nodari A, Migliavacca B, Cassetti A, Littlewood-Evans A, Reichardt LF, Messing A, Quattrini A, Mueller U, Wrabetz L. 2002. Conditional disruption of beta 1 integrin in Schwann cells impedes interactions with axons. Journal of Cell Biology 156:199–210. DOI: https://doi.org/10.1083/jcb. 200109021, PMID: 11777940 Feltri ML, Poitelon Y, Previtali SC. 2016. How Schwann cells sort axons. The Neuroscientist 22:252–265. DOI: https://doi.org/10.1177/1073858415572361 Fernandez-Valle C, Tang Y, Ricard J, Rodenas-Ruano A, Taylor A, Hackler E, Biggerstaff J, Iacovelli J. 2002. Paxillin binds schwannomin and regulates its density-dependent localization and effect on cell morphology. Nature Genetics 31:354–362. DOI: https://doi.org/10.1038/ng930, PMID: 12118253 Figlewicz DA, Quarles RH, Johnson D, Barbarash GR, Sternberger NH. 1981. Biochemical demonstration of the myelin-associated glycoprotein in the peripheral nervous system. Journal of Neurochemistry 37:749–758. DOI: https://doi.org/10.1111/j.1471-4159.1982.tb12551.x, PMID: 6168745 Fledrich R, Abdelaal T, Rasch L, Bansal V, Schu¨ tza V, Bru¨ gger B, Lu¨ chtenborg C, Prukop T, Stenzel J, Rahman RU, Hermes D, Ewers D, Mo¨ bius W, Ruhwedel T, Katona I, Weis J, Klein D, Martini R, Bru¨ ck W, Mu¨ ller WC, et al. 2018. Targeting myelin lipid metabolism as a potential therapeutic strategy in a model of CMT1A neuropathy. Nature Communications 9:3025. DOI: https://doi.org/10.1038/s41467-018-05420-0, PMID: 300726 89 Fuhrmann-Stroissnigg H, Noiges R, Descovich L, Fischer I, Albrecht DE, Nothias F, Froehner SC, Propst F. 2012. The light chains of microtubule-associated proteins MAP1A and MAP1B interact with a1-syntrophin in the central and peripheral nervous system. PLOS ONE 7:e49722. DOI: https://doi.org/10.1371/journal.pone. 0049722, PMID: 23152929 Funabashi K, Okada N, Matsuo S, Yamamoto T, Morgan BP, Okada H. 1994. Tissue distribution of complement regulatory membrane proteins in rats. Immunology 81:444–495. PMID: 7515849 Fu¨ nfschilling U, Supplie LM, Mahad D, Boretius S, Saab AS, Edgar J, Brinkmann BG, Kassmann CM, Tzvetanova ID, Mo¨ bius W, Diaz F, Meijer D, Suter U, Hamprecht B, Sereda MW, Moraes CT, Frahm J, Goebbels S, Nave KA. 2012. Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 485:517–521. DOI: https://doi.org/10.1038/nature11007, PMID: 22622581 Gallardo E, Garcı´a A, Ramo´n C, Maravı´ E, Infante J, Gasto´n I, Alonso A´ , Combarros O, De Jonghe P, Berciano J. 2009. Charcot–Marie–Tooth disease type 2J with MPZ Thr124Met mutation: clinico-electrophysiological and MRI study of a family. Journal of Neurology 256:2061–2071. DOI: https://doi.org/10.1007/s00415-009-5251-y, PMID: 19629567

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 25 of 31 Tools and resources Neuroscience

Garbern JY, Cambi F, Tang XM, Sima AA, Vallat JM, Bosch EP, Lewis R, Shy M, Sohi J, Kraft G, Chen KL, Joshi I, Leonard DG, Johnson W, Raskind W, Dlouhy SR, Pratt V, Hodes ME, Bird T, Kamholz J. 1997. Proteolipid protein is necessary in peripheral as well as central myelin. Neuron 19:205–218. DOI: https://doi.org/10.1016/ S0896-6273(00)80360-8, PMID: 9247276 Geromanos SJ, Vissers JP, Silva JC, Dorschel CA, Li GZ, Gorenstein MV, Bateman RH, Langridge JI. 2009. The detection, correlation, and comparison of peptide precursor and product ions from data independent LC-MS with data dependant LC-MS/MS. Proteomics 9:1683–1695. DOI: https://doi.org/10.1002/pmic.200800562, PMID: 19294628 Ghidinelli M, Poitelon Y, Shin YK, Ameroso D, Williamson C, Ferri C, Pellegatta M, Espino K, Mogha A, Monk K, Podini P, Taveggia C, Nave KA, Wrabetz L, Park HT, Feltri ML. 2017. Laminin 211 inhibits in Schwann cells to modulate neuregulin 1 type III-driven myelination. PLOS Biology 15:e2001408. DOI: https:// doi.org/10.1371/journal.pbio.2001408, PMID: 28636612 Giese KP, Martini R, Lemke G, Soriano P, Schachner M. 1992. Mouse P0 gene disruption leads to Hypomyelination, abnormal expression of recognition molecules, and degeneration of myelin and axons. Cell 71:565–576. DOI: https://doi.org/10.1016/0092-8674(92)90591-Y, PMID: 1384988 Gillespie CS, Sherman DL, Blair GE, Brophy PJ. 1994. Periaxin, a novel protein of myelinating Schwann cells with a possible role in axonal ensheathment. Neuron 12:497–508. DOI: https://doi.org/10.1016/0896-6273(94) 90208-9, PMID: 8155317 Gillespie CS, Sherman DL, Fleetwood-Walker SM, Cottrell DF, Tait S, Garry EM, Wallace VC, Ure J, Griffiths IR, Smith A, Brophy PJ. 2000. Peripheral demyelination and neuropathic pain behavior in periaxin-deficient mice. Neuron 26:523–531. DOI: https://doi.org/10.1016/S0896-6273(00)81184-8, PMID: 10839370 Gitik M, Liraz-Zaltsman S, Oldenborg PA, Reichert F, Rotshenker S. 2011. Myelin down-regulates myelin by and macrophages through interactions between CD47 on myelin and SIRPa (signal regulatory protein-a) on phagocytes. Journal of Neuroinflammation 8:24. DOI: https://doi.org/10.1186/1742- 2094-8-24, PMID: 21401967 Gonc¸alves AF, Dias NG, Moransard M, Correia R, Pereira JA, Witke W, Suter U, Relvas JB. 2010. Gelsolin is required for macrophage recruitment during remyelination of the peripheral nervous system. Glia 58:706–721. DOI: https://doi.org/10.1002/glia.20956, PMID: 20014276 Gonc¸alves NP, Vægter CB, Andersen H, Østergaard L, Calcutt NA, Jensen TS. 2017. Schwann cell interactions with axons and microvessels in diabetic neuropathy. Nature Reviews Neurology 13:135–147. DOI: https://doi. org/10.1038/nrneurol.2016.201, PMID: 28134254 Greenfield S, Brostoff S, Eylar EH, Morell P. 1973. Protein composition of myelin of the peripheral nervous system. Journal of Neurochemistry 20:1207–1216. DOI: https://doi.org/10.1111/j.1471-4159.1973.tb00089.x, PMID: 4697881 Grove M, Brophy PJ. 2014. FAK is required for Schwann cell spreading on immature basal Lamina to coordinate the radial sorting of peripheral axons with myelination. Journal of Neuroscience 34:13422–13434. DOI: https:// doi.org/10.1523/JNEUROSCI.1764-14.2014, PMID: 25274820 Guilbot A, Williams A, Ravise´N, Verny C, Brice A, Sherman DL, Brophy PJ, LeGuern E, Delague V, Bareil C, Me´garbane´A, Claustres M. 2001. A mutation in periaxin is responsible for CMT4F, an autosomal recessive form of Charcot-Marie-Tooth disease. Human Molecular Genetics 10:415–421. DOI: https://doi.org/10.1093/ hmg/10.4.415, PMID: 11157804 Hartline DK, Colman DR. 2007. Rapid conduction and the evolution of giant axons and myelinated fibers. Current Biology 17:R29–R35. DOI: https://doi.org/10.1016/j.cub.2006.11.042, PMID: 17208176 Hayashi A, Nakashima K, Yamagishi K, Hoshi T, Suzuki A, Baba H. 2007. Localization of annexin II in the paranodal regions and Schmidt-Lanterman incisures in the peripheral nervous system. Glia 55:1044–1052. DOI: https://doi.org/10.1002/glia.20529, PMID: 17549680 Hess A, Lansing AI. 1953. The fine structure of peripheral nerve fibers. The Anatomical Record 117:175–199. DOI: https://doi.org/10.1002/ar.1091170205, PMID: 13104987 Hulsen T, de Vlieg J, Alkema W. 2008. BioVenn - a web application for the comparison and visualization of biological lists using area-proportional venn diagrams. BMC Genomics 9:488. DOI: https://doi.org/10.1186/ 1471-2164-9-488, PMID: 18925949 Ishibashi T, Ding L, Ikenaka K, Inoue Y, Miyado K, Mekada E, Baba H. 2004. Tetraspanin protein CD9 is a novel paranodal component regulating paranodal junctional formation. Journal of Neuroscience 24:96–102. DOI: https://doi.org/10.1523/JNEUROSCI.1484-03.2004, PMID: 14715942 Ishimoto T, Ninomiya K, Inoue R, Koike M, Uchiyama Y, Mori H. 2017. Mice lacking BCAS1, a novel myelin- associated protein, display hypomyelination, -like abnormal behaviors, and upregulation of inflammatory genes in the brain. Glia 65:727–739. DOI: https://doi.org/10.1002/glia.23129, PMID: 28230289 Ivanovic A, Horresh I, Golan N, Spiegel I, Sabanay H, Frechter S, Ohno S, Terada N, Mo¨ bius W, Rosenbluth J, Brose N, Peles E. 2012. The cytoskeletal adapter protein 4.1G organizes the internodes in peripheral myelinated nerves. Journal of Cell Biology 196:337–344. DOI: https://doi.org/10.1083/jcb.201111127, PMID: 22291039 Jahn O, Tenzer S, Werner HB. 2009. Myelin proteomics: molecular anatomy of an insulating sheath. Molecular Neurobiology 40:55–72. DOI: https://doi.org/10.1007/s12035-009-8071-2, PMID: 19452287 Jha MK, Lee Y, Russell KA, Yang F, Dastgheyb RM, Deme P, Ament XH, Chen W, Liu Y, Guan Y, Polydefkis MJ, Hoke A, Haughey NJ, Rothstein JD, Morrison BM. 2020. Monocarboxylate transporter 1 in Schwann cells contributes to maintenance of sensory nerve myelination during aging. Glia 68:161–177. DOI: https://doi.org/ 10.1002/glia.23710, PMID: 31453649

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 26 of 31 Tools and resources Neuroscience

Jung M, Sommer I, Schachner M, Nave KA. 1996. Monoclonal O10 defines a conformationally sensitive cell-surface epitope of proteolipid protein (PLP): evidence that PLP misfolding underlies dysmyelination in mutant mice. Journal of Neuroscience 16:7920–7929. DOI: https://doi.org/10.1523/JNEUROSCI.16-24-07920. 1996, PMID: 8987820 Kabzinska D, Drac H, Sherman DL, Kostera-Pruszczyk A, Brophy PJ, Kochanski A, Hausmanowa-Petrusewicz I. 2006. Charcot-Marie-Tooth type 4F disease caused by S399fsx410 mutation in the PRX gene. Neurology 66:745– 747. DOI: https://doi.org/10.1212/01.wnl.0000201269.46071.35, PMID: 16534116 Kammers K, Cole RN, Tiengwe C, Ruczinski I. 2015. Detecting significant changes in protein abundance. EuPA Open Proteomics 7:11–19. DOI: https://doi.org/10.1016/j.euprot.2015.02.002, PMID: 25821719 Kangas SM, Ohlmeier S, Sormunen R, Jouhilahti EM, Peltonen S, Peltonen J, Heape AM. 2016. An approach to comprehensive genome and proteome expression analyses in Schwann cells and neurons during peripheral nerve myelin formation. Journal of Neurochemistry 138:830–844. DOI: https://doi.org/10.1111/jnc.13722, PMID: 27364987 Kim S, Maynard JC, Sasaki Y, Strickland A, Sherman DL, Brophy PJ, Burlingame AL, Milbrandt J. 2016. Schwann cell O-GlcNAc glycosylation is required for myelin maintenance and axon integrity. Journal of Neuroscience 36: 9633–9646. DOI: https://doi.org/10.1523/JNEUROSCI.1235-16.2016, PMID: 27629714 Kleopa KA, Sargiannidou I. 2015. , gap junctions and peripheral neuropathy. Neuroscience Letters 596:27–32. DOI: https://doi.org/10.1016/j.neulet.2014.10.033, PMID: 25449862 Kuharev J, Navarro P, Distler U, Jahn O, Tenzer S. 2015. In-depth evaluation of software tools for data- independent acquisition based label-free quantification. Proteomics 15:3140–3151. DOI: https://doi.org/10. 1002/pmic.201400396, PMID: 25545627 Kusch K, Uecker M, Liepold T, Mo¨ bius W, Hoffmann C, Neumann H, Werner H, Jahn O. 2017. Partial immunoblotting of 2D-Gels: a novel method to identify post-translationally modified proteins exemplified for the myelin acetylome. Proteomes 5:3. DOI: https://doi.org/10.3390/proteomes5010003 Larocca JN, Norton WT. 2006. Isolation of myelin. Current Protocols in Cell Biology 33:3.25.1–3.25.3. DOI: https://doi.org/10.1002/0471143030.cb0325s33 Le N, Nagarajan R, Wang JYT, Araki T, Schmidt RE, Milbrandt J. 2005. Analysis of congenital hypomyelinating Egr2Lo/Lo nerves identifies Sox2 as an inhibitor of Schwann cell differentiation and myelination. PNAS 102: 2596–2601. DOI: https://doi.org/10.1073/pnas.0407836102 Leal A, Berghoff C, Berghoff M, Rojas-Araya M, Ortiz C, Heuss D, Rautenstrauss B. 2014. A Costa Rican family affected with Charcot-Marie-Tooth disease due to the myelin protein zero (MPZ) p.Thr124Met mutation shares the Belgian haplotype. Revista De Biologı´a Tropical 62:1285. DOI: https://doi.org/10.15517/rbt.v62i4.13473 LeBeau JM, Liuzzi FJ, Depto AS, Vinik AI. 1994. Differential laminin gene expression in dorsal root ganglion neurons and nonneuronal cells. Experimental Neurology 127:1–8. DOI: https://doi.org/10.1006/exnr.1994. 1074, PMID: 8200428 Lee HK, Seo IA, Park HK, Park YM, Ahn KJ, Yoo YH, Park HT. 2007. Nidogen is a prosurvival and promigratory factor for adult Schwann cells. Journal of Neurochemistry 102:686–698. DOI: https://doi.org/10.1111/j.1471- 4159.2007.04580.x, PMID: 17437540 Lee Y, Morrison BM, Li Y, Lengacher S, Farah MH, Hoffman PN, Liu Y, Tsingalia A, Jin L, Zhang PW, Pellerin L, Magistretti PJ, Rothstein JD. 2012. Oligodendroglia metabolically support axons and contribute to . Nature 487:443–448. DOI: https://doi.org/10.1038/nature11314, PMID: 22801498 Lee S, Amici S, Tavori H, Zeng WM, Freeland S, Fazio S, Notterpek L. 2014. PMP22 is critical for -mediated cellular functions and for establishing lipid rafts. Journal of Neuroscience 34:16140–16152. DOI: https://doi. org/10.1523/JNEUROSCI.1908-14.2014, PMID: 25429154 Li X, Lynn BD, Olson C, Meier C, Davidson KG, Yasumura T, Rash JE, Nagy JI. 2002. Connexin29 expression, immunocytochemistry and freeze-fracture replica immunogold labelling (FRIL) in sciatic nerve. European Journal of Neuroscience 16:795–806. DOI: https://doi.org/10.1046/j.1460-9568.2002.02149.x, PMID: 12372015 Li GZ, Vissers JP, Silva JC, Golick D, Gorenstein MV, Geromanos SJ. 2009. Database searching and accounting of multiplexed precursor and product ion spectra from the data independent analysis of simple and complex peptide mixtures. Proteomics 9:1696–1719. DOI: https://doi.org/10.1002/pmic.200800564, PMID: 19294629 Lin HH, Snyder BS, Connor JR. 1990. Transferrin expression in myelinated and non-myelinated peripheral nerves. Brain Research 526:217–220. DOI: https://doi.org/10.1016/0006-8993(90)91224-5, PMID: 2257482 Liu Y, Oppenheim RW, Sugiura Y, Lin W. 2009. Abnormal development of the neuromuscular junction in Nedd4- deficient mice. Developmental Biology 330:153–166. DOI: https://doi.org/10.1016/j.ydbio.2009.03.023, PMID: 19345204 Mantuano E, Lam MS, Shibayama M, Campana WM, Gonias SL. 2015. The NMDA receptor functions independently and as an LRP1 co-receptor to promote Schwann cell survival and migration. Journal of Cell Science 128:3478–3488. DOI: https://doi.org/10.1242/jcs.173765, PMID: 26272917 Marchesi C, Milani M, Morbin M, Cesani M, Lauria G, Scaioli V, Piccolo G, Fabrizi GM, Cavallaro T, Taroni F, Pareyson D. 2010. Four novel cases of periaxin-related neuropathy and review of the literature. Neurology 75: 1830–1838. DOI: https://doi.org/10.1212/WNL.0b013e3181fd6314, PMID: 21079185 Marques S, Zeisel A, Codeluppi S, van Bruggen D, Mendanha Falca˜ o A, Xiao L, Li H, Ha¨ ring M, Hochgerner H, Romanov RA, Gyllborg D, Mun˜ oz Manchado A, La Manno G, Lo¨ nnerberg P, Floriddia EM, Rezayee F, Ernfors P, Arenas E, Hjerling-Leffler J, Harkany T, et al. 2016. heterogeneity in the mouse juvenile and adult central nervous system. Science 352:1326–1329. DOI: https://doi.org/10.1126/science.aaf6463, PMID: 27284195

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 27 of 31 Tools and resources Neuroscience

Martini R, Mohajeri MH, Kasper S, Giese KP, Schachner M. 1995. Mice doubly deficient in the genes for P0 and myelin basic protein show that both proteins contribute to the formation of the major dense line in peripheral nerve myelin. Journal of Neuroscience 15:4488–4495. DOI: https://doi.org/10.1523/JNEUROSCI.15-06-04488. 1995, PMID: 7540676 Masaki T, Matsumura K, Hirata A, Yamada H, Hase A, Arai K, Shimizu T, Yorifuji H, Motoyoshi K, Kamakura K. 2002. Expression of dystroglycan and the laminin-alpha 2 chain in the rat peripheral nerve during development. Experimental Neurology 174:109–117. DOI: https://doi.org/10.1006/exnr.2001.7856, PMID: 11869039 Matthieu JM, Costantino-Ceccarini E, Be´ny M, Reigner J. 1980. Evidence for the association of 2’,3’-cyclic- nucleotide 3’-phosphodiesterase with myelin-related membranes in peripheral nervous system. Journal of Neurochemistry 35:1345–1350. DOI: https://doi.org/10.1111/j.1471-4159.1980.tb09008.x, PMID: 6255099 Michailov GV, Sereda MW, Brinkmann BG, Fischer TM, Haug B, Birchmeier C, Role L, Lai C, Schwab MH, Nave KA. 2004. Axonal neuregulin-1 regulates myelin sheath thickness. Science 304:700–703. DOI: https://doi.org/ 10.1126/science.1095862, PMID: 15044753 Micko S, Schlaepfer WW. 1978. Protein composition of axons and myelin from rat and human peripheral nerves. Journal of Neurochemistry 30:1041–1049. DOI: https://doi.org/10.1111/j.1471-4159.1978.tb12397.x, PMID: 666897 Mikol DD, Scherer SS, Duckett SJ, Hong HL, Feldman EL. 2002. Schwann cell caveolin-1 expression increases during myelination and decreases after axotomy. Glia 38:191–199. DOI: https://doi.org/10.1002/glia.10063, PMID: 11968057 Miyamoto T, Morita K, Takemoto D, Takeuchi K, Kitano Y, Miyakawa T, Nakayama K, Okamura Y, Sasaki H, Miyachi Y, Furuse M, Tsukita S. 2005. Tight junctions in Schwann cells of peripheral myelinated axons: a lesson from claudin-19-deficient mice. The Journal of Cell Biology 169:527–565. DOI: https://doi.org/10.1083/jcb. 200501154, PMID: 15883201 Monk KR, Naylor SG, Glenn TD, Mercurio S, Perlin JR, Dominguez C, Moens CB, Talbot WS. 2009. A - coupled receptor is essential for Schwann cells to initiate myelination. Science 325:1402–1405. DOI: https://doi. org/10.1126/science.1173474, PMID: 19745155 Monk KR, Oshima K, Jo¨ rs S, Heller S, Talbot WS. 2011. Gpr126 is essential for peripheral nerve development and myelination in mammals. Development 138:2673–2680. DOI: https://doi.org/10.1242/dev.062224, PMID: 21613327 Monk KR, Feltri ML, Taveggia C. 2015. New insights on Schwann cell development. Glia 63:1376–1393. DOI: https://doi.org/10.1002/glia.22852, PMID: 25921593 Montani L, Buerki-Thurnherr T, de Faria JP, Pereira JA, Dias NG, Fernandes R, Gonc¸alves AF, Braun A, Benninger Y, Bo¨ ttcher RT, Costell M, Nave KA, Franklin RJ, Meijer D, Suter U, Relvas JB. 2014. is required for peripheral nervous system myelination. Development 141:1553–1561. DOI: https://doi.org/10.1242/dev. 101840, PMID: 24598164 Morrison BM, Lee Y, Rothstein JD. 2013. Oligodendroglia: metabolic supporters of axons. Trends in Cell Biology 23:644–651. DOI: https://doi.org/10.1016/j.tcb.2013.07.007 Morrison BM, Tsingalia A, Vidensky S, Lee Y, Jin L, Farah MH, Lengacher S, Magistretti PJ, Pellerin L, Rothstein JD. 2015. Deficiency in monocarboxylate transporter 1 (MCT1) in mice delays regeneration of peripheral nerves following sciatic nerve crush. Experimental Neurology 263:325–338. DOI: https://doi.org/10.1016/j.expneurol. 2014.10.018, PMID: 25447940 Murata M, Shibuya Y, Munemoto S, Takeuchi J, Kobayashi M, Suzuki H, Komori T. 2006. Ultrastructural localization of alpha E-catenin in the rat sciatic nerve. The Kobe Journal of Medical Sciences 52:77–84. PMID: 16855370 Myllykoski M, Eichel MA, Jung RB, Kelm S, Werner HB, Kursula P. 2018. High-affinity heterotetramer formation between the large myelin-associated glycoprotein and the dynein light chain DYNLL1. Journal of Neurochemistry 147:764–783. DOI: https://doi.org/10.1111/jnc.14598, PMID: 30261098 Nagarajan R, Le N, Mahoney H, Araki T, Milbrandt J. 2002. Deciphering peripheral nerve myelination by using Schwann cell expression profiling. PNAS 99:8998–9003. DOI: https://doi.org/10.1073/pnas.132080999, PMID: 12084938 Nave KA, Werner HB. 2014. Myelination of the nervous system: mechanisms and functions. Annual Review of Cell and Developmental Biology 30:503–533. DOI: https://doi.org/10.1146/annurev-cellbio-100913-013101, PMID: 25288117 Nawaz S, Schweitzer J, Jahn O, Werner HB. 2013. Molecular evolution of myelin basic protein, an abundant structural myelin component. Glia 61:1364–1377. DOI: https://doi.org/10.1002/glia.22520, PMID: 24040667 Neilson KA, Ali NA, Muralidharan S, Mirzaei M, Mariani M, Assadourian G, Lee A, van Sluyter SC, Haynes PA. 2011. Less label, more free: approaches in label-free quantitative mass spectrometry. Proteomics 11:535–553. DOI: https://doi.org/10.1002/pmic.201000553, PMID: 21243637 Nodari A, Previtali SC, Dati G, Occhi S, Court FA, Colombelli C, Zambroni D, Dina G, Del Carro U, Campbell KP, Quattrini A, Wrabetz L, Feltri ML. 2008. Alpha6 beta4 integrin and dystroglycan cooperate to stabilize the myelin sheath. Journal of Neuroscience 28:6714–6719. DOI: https://doi.org/10.1523/JNEUROSCI.0326-08. 2008, PMID: 18579745 Ogata T, Iijima S, Hoshikawa S, Miura T, Yamamoto S, Oda H, Nakamura K, Tanaka S. 2004. Opposing extracellular signal-regulated kinase and akt pathways control Schwann cell myelination. Journal of Neuroscience 24:6724–6732. DOI: https://doi.org/10.1523/JNEUROSCI.5520-03.2004, PMID: 15282275

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 28 of 31 Tools and resources Neuroscience

Ohno N, Terada N, Yamakawa H, Komada M, Ohara O, Trapp BD, Ohno S. 2006. Expression of protein 4.1G in Schwann cells of the peripheral nervous system. Journal of Neuroscience Research 84:568–577. DOI: https:// doi.org/10.1002/jnr.20949, PMID: 16752423 Pareyson D, Marchesi C. 2009. Diagnosis, natural history, and management of Charcot-Marie-Tooth disease. The Lancet Neurology 8:654–667. DOI: https://doi.org/10.1016/S1474-4422(09)70110-3, PMID: 19539237 Patzig J, Jahn O, Tenzer S, Wichert SP, de Monasterio-Schrader P, Rosfa S, Kuharev J, Yan K, Bormuth I, Bremer J, Aguzzi A, Orfaniotou F, Hesse D, Schwab MH, Mo¨ bius W, Nave KA, Werner HB. 2011. Quantitative and integrative proteome analysis of peripheral nerve myelin identifies novel myelin proteins and candidate neuropathy loci. Journal of Neuroscience 31:16369–16386. DOI: https://doi.org/10.1523/JNEUROSCI.4016-11. 2011, PMID: 22072688 Patzig J, Kusch K, Fledrich R, Eichel MA, Lu¨ ders KA, Mo¨ bius W, Sereda MW, Nave K-A, Martini R, Werner HB. 2016a. Proteolipid protein modulates preservation of peripheral axons and premature death when myelin protein zero is lacking. Glia 64:155–174. DOI: https://doi.org/10.1002/glia.22922 Patzig J, Erwig MS, Tenzer S, Kusch K, Dibaj P, Mo¨ bius W, Goebbels S, Schaeren-Wiemers N, Nave KA, Werner HB. 2016b. Septin/anillin filaments scaffold central nervous system myelin to accelerate nerve conduction. eLife 5:e17119. DOI: https://doi.org/10.7554/eLife.17119, PMID: 27504968 Pereira JA, Lebrun-Julien F, Suter U. 2012. Molecular mechanisms regulating myelination in the peripheral nervous system. Trends in Neurosciences 35:123–134. DOI: https://doi.org/10.1016/j.tins.2011.11.006, PMID: 22192173 Petersen SC, Luo R, Liebscher I, Giera S, Jeong SJ, Mogha A, Ghidinelli M, Feltri ML, Scho¨ neberg T, Piao X, Monk KR. 2015. The adhesion GPCR GPR126 has distinct, domain-dependent functions in Schwann cell development mediated by interaction with laminin-211. Neuron 85:755–769. DOI: https://doi.org/10.1016/j. neuron.2014.12.057, PMID: 25695270 Philp NJ, Ochrietor JD, Rudoy C, Muramatsu T, Linser PJ. 2003. Loss of MCT1, MCT3, and MCT4 expression in the retinal pigment and neural of the 5a11/Basigin-Null mouse. Investigative Opthalmology & Visual Science 44:1305. DOI: https://doi.org/10.1167/iovs.02-0552 Poliak S, Matlis S, Ullmer C, Scherer SS, Peles E. 2002. Distinct and associated PDZ proteins form different autotypic tight junctions in myelinating Schwann cells. Journal of Cell Biology 159:361–372. DOI: https://doi.org/10.1083/jcb.200207050, PMID: 12403818 Quattrini A, Previtali S, Feltri ML, Canal N, Nemni R, Wrabetz L. 1996. Beta 4 integrin and other Schwann cell markers in axonal neuropathy. Glia 17:294–306. DOI: https://doi.org/10.1002/(SICI)1098-1136(199608)17: 4<294::AID-GLIA4>3.0.CO;2-#, PMID: 8856326 Raasakka A, Linxweiler H, Brophy PJ, Sherman DL, Kursula P. 2019. Direct binding of the flexible C-Terminal segment of periaxin to b4 integrin suggests a molecular basis for CMT4F. Frontiers in Molecular Neuroscience 12:84. DOI: https://doi.org/10.3389/fnmol.2019.00084 Rende M, Muir D, Ruoslahti E, Hagg T, Varon S, Manthorpe M. 1992. Immunolocalization of ciliary neuronotrophic factor in adult rat sciatic nerve. Glia 5:25–32. DOI: https://doi.org/10.1002/glia.440050105, PMID: 1531807 Riethmacher D, Sonnenberg-Riethmacher E, Brinkmann V, Yamaai T, Lewin GR, Birchmeier C. 1997. Severe neuropathies in mice with targeted mutations in the ErbB3 receptor. Nature 389:725–730. DOI: https://doi. org/10.1038/39593, PMID: 9338783 Rinholm JE, Bergersen LH. 2014. White matter lactate–does it matter? Neuroscience 276:109–116. DOI: https:// doi.org/10.1016/j.neuroscience.2013.10.002, PMID: 24125892 Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. 2015. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Research 43:e47. DOI: https://doi.org/10. 1093/nar/gkv007, PMID: 25605792 Rossor AM, Polke JM, Houlden H, Reilly MM. 2013. Clinical implications of genetic advances in Charcot-Marie- Tooth disease. Nature Reviews Neurology 9:562–571. DOI: https://doi.org/10.1038/nrneurol.2013.179, PMID: 24018473 Ryu EJ, Yang M, Gustin JA, Chang LW, Freimuth RR, Nagarajan R, Milbrandt J. 2008. Analysis of peripheral nerve expression profiles identifies a novel myelin glycoprotein, MP11. Journal of Neuroscience 28:7563–7573. DOI: https://doi.org/10.1523/JNEUROSCI.1659-08.2008, PMID: 18650334 Saab AS, Tzvetanova ID, Nave KA. 2013. The role of myelin and oligodendrocytes in axonal energy metabolism. Current Opinion in Neurobiology 23:1065–1072. DOI: https://doi.org/10.1016/j.conb.2013.09.008, PMID: 240 94633 Saitoh Y, Ohno N, Yamauchi J, Sakamoto T, Terada N. 2017. Deficiency of a membrane skeletal protein, 4.1G, results in myelin abnormalities in the peripheral nervous system. Histochemistry and Cell Biology 148:597–606. DOI: https://doi.org/10.1007/s00418-017-1600-6, PMID: 28755316 Saitoh Y, Kamijo A, Yamauchi J, Sakamoto T, Terada N. 2019. The membrane palmitoylated protein, MPP6, is involved in myelin formation in the mouse peripheral nervous system. Histochemistry and Cell Biology 151: 385–394. DOI: https://doi.org/10.1007/s00418-018-1745-y, PMID: 30357511 Salles J, Sargueil F, Knoll-Gellida A, Witters LA, Shy M, Jiang H, Cassagne C, Garbay B. 2002. Fatty acid synthase expression during peripheral nervous system myelination. Molecular Brain Research 101:52–58. DOI: https:// doi.org/10.1016/S0169-328X(02)00161-4, PMID: 12007831 Schaeren-Wiemers N, Bonnet A, Erb M, Erne B, Bartsch U, Kern F, Mantei N, Sherman D, Suter U. 2004. The raft-associated protein MAL is required for maintenance of proper axon–glia interactions in the central nervous system. Journal of Cell Biology 166:731–742. DOI: https://doi.org/10.1083/jcb.200406092, PMID: 15337780

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 29 of 31 Tools and resources Neuroscience

Schardt A, Brinkmann BG, Mitkovski M, Sereda MW, Werner HB, Nave KA. 2009. The SNARE protein SNAP-29 interacts with the GTPase Rab3A: implications for membrane trafficking in myelinating Glia. Journal of Neuroscience Research 87:3465–3479. DOI: https://doi.org/10.1002/jnr.22005, PMID: 19170188 Scheiermann C, Meda P, Aurrand-Lions M, Madani R, Yiangou Y, Coffey P, Salt TE, Ducrest-Gay D, Caille D, Howell O, Reynolds R, Lobrinus A, Adams RH, Yu AS, Anand P, Imhof BA, Nourshargh S. 2007. Expression and function of junctional adhesion molecule-C in myelinated peripheral nerves. Science 318:1472–1475. DOI: https://doi.org/10.1126/science.1149276, PMID: 18048693 Scherer SS, Xu T, Crino P, Arroyo EJ, Gutmann DH. 2001. Ezrin, radixin, and moesin are components of Schwann cell microvilli. Journal of Neuroscience Research 65:150–164. DOI: https://doi.org/10.1002/jnr.1138, PMID: 11438984 Sharma K, Schmitt S, Bergner CG, Tyanova S, Kannaiyan N, Manrique-Hoyos N, Kongi K, Cantuti L, Hanisch UK, Philips MA, Rossner MJ, Mann M, Simons M. 2015. Cell type- and brain region-resolved mouse brain proteome. Nature Neuroscience 18:1819–1831. DOI: https://doi.org/10.1038/nn.4160, PMID: 26523646 Sherman DL, Fabrizi C, Gillespie CS, Brophy PJ. 2001. Specific disruption of a Schwann cell dystrophin-related protein complex in a demyelinating neuropathy. Neuron 30:677–687. DOI: https://doi.org/10.1016/S0896-6273 (01)00327-0, PMID: 11430802 Sherman DL, Brophy PJ. 2005. Mechanisms of axon ensheathment and myelin growth. Nature Reviews Neuroscience 6:683–690. DOI: https://doi.org/10.1038/nrn1743, PMID: 16136172 Shih IM, Nesbit M, Herlyn M, Kurman RJ. 1998. A new Mel-CAM (CD146)-specific monoclonal antibody, MN-4, on paraffin-embedded tissue. Modern Pathology 11:1098–1204. PMID: 9831208 Silva JC, Gorenstein MV, Li G-Z, Vissers JPC, Geromanos SJ. 2006. Absolute quantification of proteins by LCMS E. Molecular & Cellular Proteomics 5:144–156. DOI: https://doi.org/10.1074/mcp.M500230-MCP200 Smith ME, Curtis BM. 1979. Frog sciatic nerve myelin: a chemical characterization. Journal of Neurochemistry 33: 447–452. DOI: https://doi.org/10.1111/j.1471-4159.1979.tb05174.x, PMID: 112219 Sparrow N, Manetti ME, Bott M, Fabianac T, Petrilli A, Bates ML, Bunge MB, Lambert S, Fernandez-Valle C. 2012. The actin-severing protein cofilin is downstream of neuregulin signaling and is essential for Schwann cell myelination. Journal of Neuroscience 32:5284–5297. DOI: https://doi.org/10.1523/JNEUROSCI.6207-11.2012, PMID: 22496574 Spiegel I, Adamsky K, Eshed Y, Milo R, Sabanay H, Sarig-Nadir O, Horresh I, Scherer SS, Rasband MN, Peles E. 2007. A central role for Necl4 (SynCAM4) in schwann cell-axon interaction and myelination. Nature Neuroscience 10:861–869. DOI: https://doi.org/10.1038/nn1915, PMID: 17558405 Stassart RM, Mo¨ bius W, Nave K-A, Edgar JM. 2018. The axon-myelin unit in development and degenerative disease. Frontiers in Neuroscience 12:00467. DOI: https://doi.org/10.3389/fnins.2018.00467 Storey JD. 2003. The positive false discovery rate: a Bayesian interpretation and the q -value. The Annals of Statistics 31:2013–2035. DOI: https://doi.org/10.1214/aos/1074290335 Stumpf SK, Berghoff SA, Trevisiol A, Spieth L, Du¨ king T, Schneider LV, Schlaphoff L, Dreha-Kulaczewski S, Bley A, Burfeind D, Kusch K, Mitkovski M, Ruhwedel T, Guder P, Ro¨ hse H, Denecke J, Ga¨ rtner J, Mo¨ bius W, Nave KA, Saher G. 2019. Ketogenic diet ameliorates axonal defects and promotes myelination in Pelizaeus- Merzbacher disease. Acta Neuropathologica 138:147–161. DOI: https://doi.org/10.1007/s00401-019-01985-2, PMID: 30919030 Susuki K, Zollinger DR, Chang KJ, Zhang C, Huang CY, Tsai CR, Galiano MR, Liu Y, Benusa SD, Yermakov LM, Griggs RB, Dupree JL, Rasband MN. 2018. Glial bii spectrin contributes to paranode formation and maintenance. Journal of Neuroscience 38:6063–6075. DOI: https://doi.org/10.1523/JNEUROSCI.3647-17.2018, PMID: 29853631 Tait S, Gunn-Moore F, Collinson JM, Huang J, Lubetzki C, Pedraza L, Sherman DL, Colman DR, Brophy PJ. 2000. An oligodendrocyte cell adhesion molecule at the site of assembly of the paranodal axo-glial junction. The Journal of Cell Biology 150:657–666. DOI: https://doi.org/10.1083/jcb.150.3.657, PMID: 10931875 Taveggia C, Zanazzi G, Petrylak A, Yano H, Rosenbluth J, Einheber S, Xu X, Esper RM, Loeb JA, Shrager P, Chao MV, Falls DL, Role L, Salzer JL. 2005. Neuregulin-1 type III determines the ensheathment fate of axons. Neuron 47:681–694. DOI: https://doi.org/10.1016/j.neuron.2005.08.017, PMID: 16129398 Thakurela S, Garding A, Jung RB, Mu¨ ller C, Goebbels S, White R, Werner HB, Tiwari VK. 2016. The transcriptome of mouse central nervous system myelin. Scientific Reports 6:25828. DOI: https://doi.org/10. 1038/srep25828, PMID: 27173133 Tokuda N, Noto Y, Kitani-Morii F, Hamano A, Kasai T, Shiga K, Mizuta I, Niwa F, Nakagawa M, Mizuno T. 2015. Parasympathetic dominant autonomic dysfunction in Charcot-Marie-Tooth disease type 2J with the MPZ Thr124Met Mutation. Internal Medicine 54:1919–1922. DOI: https://doi.org/10.2169/internalmedicine.54.4259, PMID: 26234237 Tokunaga S, Hashiguchi A, Yoshimura A, Maeda K, Suzuki T, Haruki H, Nakamura T, Okamoto Y, Takashima H. 2012. Late-onset Charcot-Marie-Tooth disease 4F caused by periaxin gene mutation. Neurogenetics 13:359– 365. DOI: https://doi.org/10.1007/s10048-012-0338-5, PMID: 22847150 Trapp BD, Dubois-Dalcq M, Quarles RH. 1984. Ultrastructural localization of P2 protein in actively myelinating rat Schwann cells. Journal of Neurochemistry 43:944–952. DOI: https://doi.org/10.1111/j.1471-4159.1984.tb12828. x, PMID: 6206203 Triolo D, Dina G, Taveggia C, Vaccari I, Porrello E, Rivellini C, Domi T, La Marca R, Cerri F, Bolino A, Quattrini A, Previtali SC. 2012. Vimentin regulates peripheral nerve myelination. Development 139:1359–1367. DOI: https:// doi.org/10.1242/dev.072371, PMID: 22357929

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 30 of 31 Tools and resources Neuroscience

Verheijen MH, Chrast R, Burrola P, Lemke G. 2003. Local regulation of fat metabolism in peripheral nerves. Genes & Development 17:2450–2464. DOI: https://doi.org/10.1101/gad.1116203, PMID: 14522948 Vizcaı´noJA, Csordas A, del-Toro N, Dianes JA, Griss J, Lavidas I, Mayer G, Perez-Riverol Y, Reisinger F, Ternent T, Xu QW, Wang R, Hermjakob H. 2016. Update of the PRIDE database and its related tools. Nucleic Acids Research 44:D447–D456. DOI: https://doi.org/10.1093/nar/gkv1145, PMID: 26527722 Weil MT, Heibeck S, To¨ pperwien M, Tom Dieck S, Ruhwedel T, Salditt T, Rodicio MC, Morgan JR, Nave KA, Mo¨ bius W, Werner HB. 2018. Axonal ensheathment in the nervous system of lamprey: implications for the evolution of myelinating glia. Journal of Neuroscience 38:6586–6596. DOI: https://doi.org/10.1523/ JNEUROSCI.1034-18.2018, PMID: 29941446 Werner HB, Kuhlmann K, Shen S, Uecker M, Schardt A, Dimova K, Orfaniotou F, Dhaunchak A, Brinkmann BG, Mo¨ bius W, Guarente L, Casaccia-Bonnefil P, Jahn O, Nave KA. 2007. Proteolipid protein is required for transport of sirtuin 2 into CNS myelin. Journal of Neuroscience 27:7717–7730. DOI: https://doi.org/10.1523/ JNEUROSCI.1254-07.2007, PMID: 17634366 Whitaker JN. 1981. The protein antigens of peripheral nerve myelin. Annals of Neurology 9:56–64. DOI: https:// doi.org/10.1002/ana.410090710, PMID: 6164338 Woldeyesus MT, Britsch S, Riethmacher D, Xu L, Sonnenberg-Riethmacher E, Abou-Rebyeh F, Harvey R, Caroni P, Birchmeier C. 1999. Peripheral nervous system defects in erbB2 mutants following genetic rescue of heart development. Genes & Development 13:2538–2548. DOI: https://doi.org/10.1101/gad.13.19.2538, PMID: 10521398 Yamada H, Shimizu T, Tanaka T, Campbell KP, Matsumura K. 1994. Dystroglycan is a binding protein of laminin and merosin in peripheral nerve. FEBS Letters 352:49–53. DOI: https://doi.org/10.1016/0014-5793(94)00917-1, PMID: 7925941 Yang D, Bierman J, Tarumi YS, Zhong YP, Rangwala R, Proctor TM, Miyagoe-Suzuki Y, Takeda S, Miner JH, Sherman LS, Gold BG, Patton BL. 2005. Coordinate control of axon defasciculation and myelination by laminin- 2 and -8. Journal of Cell Biology 168:655–666. DOI: https://doi.org/10.1083/jcb.200411158, PMID: 15699217 Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, Phatnani HP, Guarnieri P, Caneda C, Ruderisch N, Deng S, Liddelow SA, Zhang C, Daneman R, Maniatis T, Barres BA, Wu JQ. 2014. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. Journal of Neuroscience 34:11929–11947. DOI: https://doi.org/10.1523/JNEUROSCI.1860-14.2014, PMID: 25186741

Siems et al. eLife 2020;9:e51406. DOI: https://doi.org/10.7554/eLife.51406 31 of 31